Quantcast
Channel: DR ANTHONY MELVIN CRASTO Ph.D – New Drug Approvals
Viewing all 1676 articles
Browse latest View live

 JTV 519, K 201, 

$
0
0

JTV-519.svg

JTV-519

  • Molecular FormulaC25H32N2O2S
  • Average mass424.599 Da
  • 145903-06-6 CAS

ChemSpider 2D Image | JTV-519 hydrochloride salt | C25H33ClN2O2S

JTV-519 hydrochloride salt

  • Molecular FormulaC25H33ClN2O2S
  • Average mass461.060 Da
3-(4-Benzyl-1-piperidinyl)-1-(7-methoxy-2,3-dihydro-1,4-benzothiazepin-4(5H)-yl)-1-propanonhydrochlorid (1:1)
4-[3-(4-benzylpiperidin-1-yl)propanoyl]-7-methoxy-2,3,4,5-tetrahydro-1,4-benzothiazepine hydrochloride
JTV-519 hydrochloride salt
1038410-88-6 [RN]
  1. UNII-0I621Y6R4Q
  2. K201
  3. 1038410-88-6
  4. K 201
  5. SCHEMBL194018
  6. CHEMBL2440857
  7. DTXSID90146108
  8. 0I621Y6R4Q
  9. LS-193564

Image result for Andrew Marks, JAPAN TOBACCO

JAPAN TOBACCO

Acute Myocardial Infarction, Treatment of Cardiovascular Diseases (Not Specified)
Antiarrhythmic Drugs

JTV-519 (K201) is a 1,4-benzothiazepine derivative that interacts with many cellular targets.[1] It has many structural similarities to diltiazem, a Ca2+ channel blocker used for treatment of hypertensionangina pectoris and some types of arrhythmias.[2] JTV-519 acts in the sarcoplasmic reticulum (SR) of cardiac myocytes by binding to and stabilizing the ryanodine receptor (RyR2) in its closed state.[3][4]It can be used in the treatment of cardiac arrhythmias, heart failurecatecholaminergic polymorphic ventricular tachycardia (CPVT) and store overload-induced Ca2+ release (SOICR).[2][3][4] Currently, this drug has only been tested on animals and its side effects are still unknown.[5] As research continues, some studies have also found a dose-dependent response; where there is no improvement seen in failing hearts at 0.3 μM and a decline in response at 1 μM.[4]

K-201 (JTV-519; 1,4-benzothiazepine derivative) is an antiarrhythmic drug, had been in phase II clinical development at Japan Tobacco and Sequel Pharmaceuticals for the intravenous treatment of atrial fibrillation; however no recent developments have been reported and Sequel Pharmaceuticals has ceased operations.

In 2006, NovaCardia acquired rights from Aetas to develop the product in Europe and US for cardiovascular disorders. Sequel acquired the compound, which has a unique multi-ion channel profile, from NovaCardia following its acquisition by Merck & Co.

Treatment with JTV-519 involves stabilization of RyR2 in its closed state, decreasing its open probability during diastole and inhibiting a Ca2+ leak into the cell’s cytosol.[3][4] By decreasing the intracellular Ca2+ leak, it is able to prevent Ca2+ sparks or increases in the resting membrane potential, which can lead to spontaneous depolarization (cardiac arrhythmias), and eventually heart failure, due to the unsynchronized contraction of the atrial and ventricular compartments of the heart.[2][3][4] When Ca2+ sparks occur from the SR, the increase in intracellular Ca2+ contributes to the rising membrane potential which leads to the irregular heart beat associated to cardiac arrhythmias.[3] It can also prevent SOICR in the same manner; preventing opening of the channel due to the increase of Ca2+ inside the SR levels beyond its threshold.[2]

Molecular problem

In the closed state, N-terminal and central domains come into close contact interacting to cause a “zipping” of domains. This leads to conformational constraints that stabilize the channel and maintain the closed state.[1] Most RyR2 mutations are clustered into three regions of the channel, all affecting the same domains that interact to stabilize the channel.[1] Any of these mutations can lead to “unzipping” of the domains and a decrease in the energy barrier required for opening the channel (increasing its open probability).[1]This channel “unzipping” allows for an increase in protein kinase A phosphorylation and calstabin2 dissociation. Phosphorylation of RyR2 increases the channel’s response to Ca2+, which usually binds the RyR2 to open it.[1] If the channel become phosphorylated, this can lead to an increase in Ca2+ sparks due to an increase in Ca2+ sensitivity.

Some researchers believe that the depletion of calstabin2 from the RyR2 causes the calcium leak.[3] The depletion of calstabin2 can occur in both heart failure and CPVT.[3]Calstabin2 is a protein that stabilizes RyR2 in its closed state, preventing Ca2+ leakage during diastole. When calstabin2 is lost, the interdomain interactions of RyR2 become loose, allowing the Ca2+ leak.[3] However, the role of calstabin2 has been controversial, as some studies have found it necessary for the effect of JTV-519,[3] whereas others have found the drug functions without the stabilizing protein.[2]

Molecular mechanism

JTV-519 seems to restore the stable conformation of RyR2 during the closed state.[1][4] It is still controversial whether or not calstabin2 is necessary for this process, however, many studies believe that JTV-519 can act directly on the channel and by binding, prevents conformational changes.[2] This stabilization of the channel decreases its open probability resulting in fewer leaks of Ca2+ into the cytosol and fewer Ca2+ sparks to occur.[3][4] Researchers who believe that calstabin2 is necessary for JTV-519 effect, found that this drug may function by inducing the binding of calstabin2 back to the channel or increasing calstabin2’s affinity for the RyR2 and thus increasing its stability.[2][3]

SYNTHESIS

PATENT

US 20050186640

https://www.google.com/patents/US20050186640

Inventors Andrew MarksDonald LandryShi DengZhen Cheng
Original Assignee Marks Andrew R.Landry Donald W.Deng Shi X.Cheng Zhen Z.

PATENT

WO 9212148

https://www.google.co.in/patents/WO1992012148A1?cl=en

Inventors Noboru KanekoTatsushi OosawaTeruyuki SakaiHideo Oota
Applicant Noboru Kaneko

PATENT

US 2014121368

2,3,4,5-tetrahydrobenzo[f][1,4]thiazepines are important compounds because of their biological activities, as disclosed, for example, in U.S. Pat. Nos. 5,416,066 and 5,580,866 and published US Patent Applications Nos. 2005/0215540, 2007/0049572 and 2007/0173482.

Synthetic procedures exist for the preparation of 2-oxo-, 3-oxo-, 5-oxo- and 3,5-dioxo-1,4-benzothiazepines and for 2,3-dihydro-1,4-benzothiazepines. However, relatively few publications describe the preparation of 2,3,4,5-tetrahydrobenzo-1,4-thiazepines that contain no carbonyl groups, and most of these involve reduction of a carbonyl group or an imine. Many of the routes described in the literature proceed from an ortho-substituted arene and use the ortho substituents as “anchors” for the attachment of the seven-membered ring. Essentially all the preparatively useful syntheses in the literature that do not begin with an ortho-substituted arene employ a modification of the Bischler-Napieralski reaction in which the carbon of the acyl group on the γ-amide becomes the carbon adjacent the bridgehead and the acyl substituent becomes the 5-substituent. Like earlier mentioned syntheses, the Bischler-Napieralski synthesis requires reduction of an iminium intermediate.

It would be useful to have an intramolecular reaction for the direct construction of 2,3,4,5-tetrahydrobenzo[1,4]thiazepines that would allow more flexibility in the 4- and 5-substituents and that would avoid a separate reduction step. The Pictet Spengler reaction, in which a β-arylethylamine such as tryptamine undergoes 6-membered ring closure after condensation (cyclization) with an aldehyde, has been widely used in the synthesis of 6-membered ring systems over the past century and might be contemplated for this purpose. The Pictet Spengler reaction, however, has not been generally useful for 7-membered ring systems such as 1,4-benzothiazepines. A plausible explanation is that the failure of the reaction for typical arenes was due to the unfavorable conformation of the 7-membered ring. There are two isolated examples of an intramolecular Pictet-Spengler-type reaction producing a good yield of a benzothiazepine from the addition of formaldehyde. In one case, the starting material was a highly unusual activated arene (a catechol derivative) [Manini et al. J. Org. Chem. (2000), 65, 4269-4273]. In the other case, the starting material is a bis(benzotriazolylmethyl)amine that cyclizes to a mono(benzotriazolyl)benzothiazole [Katritzky et al. J. Chem. Soc. Pl (2002), 592-598].

PATENT

US 20050186640

WO 2015031914

US 20040229781

US 20090292119

US 7704990

PAPER

Journal of Medicinal Chemistry (2013), 56(21), 8626-8655

http://pubs.acs.org/doi/full/10.1021/jm401090a

PAPER

Synthesis of 2,3,4,5-Tetrahydrobenzo[1,4]thiazepines via N-Acyliminium Cyclization

 ARMGO Pharma, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, United States
 Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00260
Publication Date (Web): September 28, 2017
Copyright © 2017 American Chemical Society
*Phone: (914)-425-0000. E-mail: sbelvedere@armgo.com.

Abstract

Abstract Image

We report an efficient and scalable synthesis of 7-methoxy-2,3,4,5-tetrahydrobenzo[1,4]thiazepine, the core structure of biologically active molecules like JTV-519 and S107. This synthetic route, starting with 4-methoxythiophenol and proceeding via acyliminum cyclization, gives the target product in four steps and 68% overall yield and is a substantial improvement over previously published processes. Nine additional examples of tetrahydrobenzo[1,4]thiazepine synthesis via acyliminium ring closure are also presented.

References

  1. Jump up to:a b c d e f Oda, T; Yano, M; Yamamoto, T; Tokuhisa, T; Okuda, S; Doi, M; Ohkusa, T; Ikeda, Y; et al. (2005). “Defective regulation of interdomain interactions within the ryanodine receptor plays a key role in the pathogenesis of heart failure”. Circulation111 (25): 3400–10. PMID 15967847doi:10.1161/CIRCULATIONAHA.104.507921.
  2. Jump up to:a b c d e f g Hunt, DJ; Jones, PP; Wang, R; Chen, W; Bolstad, J; Chen, K; Shimoni, Y; Chen, SR (2007). “K201 (JTV519) suppresses spontaneous Ca2+ release and 3Hryanodine binding to RyR2 irrespective of FKBP12.6 association”The Biochemical Journal404 (3): 431–8. PMC 1896290Freely accessiblePMID 17313373doi:10.1042/BJ20070135.
  3. Jump up to:a b c d e f g h i j k Wehrens, XH; Lehnart, SE; Reiken, SR; Deng, SX; Vest, JA; Cervantes, D; Coromilas, J; Landry, DW; Marks, AR (2004). “Protection from cardiac arrhythmia through ryanodine receptor-stabilizing protein calstabin2”. Science304 (5668): 292–6. PMID 15073377doi:10.1126/science.1094301.
  4. Jump up to:a b c d e f g Toischer, K; Lehnart, SE; Tenderich, G; Milting, H; Körfer, R; Schmitto, JD; Schöndube, FA; Kaneko, N; et al. (2010). “K201 improves aspects of the contractile performance of human failing myocardium via reduction in Ca2+ leak from the sarcoplasmic reticulum”Basic research in cardiology105 (2): 279–87. PMC 2807967Freely accessiblePMID 19718543doi:10.1007/s00395-009-0057-8.
  5. Jump up^ Viswanathan, MN; Page, RL (2009). “Pharmacological therapy for atrial fibrillation: Current options and new agents”. Expert Opinion on Investigational Drugs18 (4): 417–31. PMID 19278302doi:10.1517/13543780902773410.
JTV-519
JTV-519.svg
Names
IUPAC name
3-(4-Benzyl-1-piperidinyl)-1-(7-methoxy-2,3-dihydro-1,4-benzothiazepin-4(5H)-yl)-1-propanone
Other names
K201
Identifiers
3D model (JSmol)
ChemSpider
PubChem CID
UNII
Properties
C25H32N2O2S
Molar mass 424.60 g·mol−1
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

//////////////JTV-519K201, JTV 519, K 201, 


Filed under: Uncategorized Tagged: JTV-519, K 201, K201

FDA clears first 7T magnetic resonance imaging device

Synthesis of isosorbide: an overview of challenging reactions

$
0
0
Green Chem., 2017, Advance Article
DOI: 10.1039/C7GC01912B, Tutorial Review
C. Dussenne, T. Delaunay, V. Wiatz, H. Wyart, I. Suisse, M. Sauthier
This review gives an overview of the catalysts and technologies developed for the synthesis of isosorbide, a platform molecule derived from biomass (sorbitol and cellulose).

Synthesis of isosorbide: an overview of challenging reactions

 Author affiliations

Abstract

Isosorbide is a diol derived from sorbitol and obtained through dehydration reactions that has raised much interest in the literature over the past few decades. Thus, this platform chemical is a biobased alternative to a number of petrosourced molecules that can find applications in a large number of technical specialty fields, such as plasticizers, monomers, solvents or pharmaceuticals. The synthesis of isosorbide is still a technical challenge, as several competitive reactions must be simultaneously handled to promote a high molar yield and avoid side reactions, like degradation and polymerization. In this purpose, many studies have proposed innovative and varied methods with promising results. This review gives an overview of the synthesis strategies and catalysts developed to access this very attractive molecule, pointing out both the results obtained and the remaining issues connected to isosorbide synthesis.

STR1 STR2

Up to now, isosorbide has been used to access a large panel of molecules with relevant applicative properties and industrial reality (Scheme 2).12 Isosorbide dinitrate is used since several decades as vasodilator.13, 14 The dimethyl isosorbide is for example used as solvent in cosmetics15-17 and isosorbide diesters18-22 are actually industrially produced and commercialized as surfactants23-27 and PVC plasticizer28, 29 . The rigid scaffold associated to the bifunctionality of the molecule has attracted a strong interest in the field of polymers chemistry. Isosorbide and derivatives have thus been shown as suitable monomers for the industrial production of polycarbonates30, 31, polyesters32-41 or polyamides42-44, with attractive applicative properties. For example, isosorbide allows the increase of Tg, improves the scratch resistance and gives excellent optical properties to polymers. Polyesters and polycarbonates containing isosorbide have now commercial developments in food packaging, spray container, automotive, material for electronic devices … .

Conclusions

Isosorbide is a versatile platform molecule that shows key features to make it a credible alternative to petro-based products. The molecule is already available on large industrial scale (tens of thousands tons per years), which allows its development in commercial products such as active pharma ingredient, additive for cosmetic, speciality chemicals and polymers (ex: polycarbonates – polyesters). The development of more selective and higher yields syntheses of isosorbide are greatly needed to consolidate isosorbide production in view of a large expansion of its uses. Sorbitol conversion to isosorbide, relying on a starch route, is already a tough challenge. In a farther future, development of a credible path to isosorbide relying on cellulose source could even be thought of, provided that very versatile innovative catalysts will be developed in the next years. In all cases, a key issue is to develop catalysts that will avoid the massive production of “oligomeric/polymeric” by-products in order to access more sustainable processes by limiting the amounts of wastes produced during the synthesis. For this purpose, more selective homogeneous catalysts than the conventional Brønsted acids or alternative reaction conditions would be of strong interest. Selective and recyclable heterogeneous catalysts would be even more profitable as they would allow the continuous production of catalyst free isosorbide. This latter approach faces strong limitations due to the need of high reaction temperatures that often result in high amounts of side-products and the need of frequent and often tedious catalyst regeneration. Innovation concerning isosorbide synthesis is still an open field on which the design of efficient and robust catalysts, either homogeneous or heterogeneous, is a key issue. Such developments would pave the way to high scale effective processes considering altogether synthesis and purification of isosorbide.

////////////////

Image result for ISOSORBIDE SYNTHESIS

Image result for ISOSORBIDE SYNTHESIS

Isosorbide is a heterocyclic compound that is derived from glucose. Isosorbide and its two isomers, namely isoidide and isomannide, are 1,4:3,6-dianhydrohexitols. It is a white solid that is prepared from the double dehydration of sorbitol. Isosorbide is a non-toxic diolproduced from biobased feedstocks, that is biodegradable and thermally stable. It is used in medicine and has been touted as a potential biofeedstock.

Production

Hydrogenation of glucose gives sorbitol. Isosorbide is obtained by double dehydration of sorbitol:

(CHOH)4(CH2OH)2 → C6H10O2(OH)2 + 2 H2O

An intermediate in the dehydration is the monocycle sorbitan.[1]

Application

Isosorbide is used as a diuretic, mainly to treat hydrocephalus, and is also used to treat glaucoma.[2] Other medications are derived from isosorbide, including isosorbide dinitrate and isosorbide mononitrate, are used to treat angina pectoris. Other isosorbide-based medicines are used as osmotic diuretics and for treatment of esophageal varices. Like other nitric oxide donors (see biological functions of nitric oxide), these drugs lower portal pressure by vasodilation and decreasing cardiac output. Isosorbide dinitrate and hydralazineare the two components of the anti-hypertensive drug isosorbide dinitrate/hydralazine (Bidil).

Isosorbide is also used as a building block for bio based polymers such as polyesters.[3]

References

  1. Jump up^ M. Rose, R. Palkovits (2012). “Isosorbide as a Renewable Platform chemical for Versatile Applications—Quo Vadis?”. ChemSusChem5 (1): 167–176. PMID 22213713doi:10.1002/cssc.201100580.
  2. Jump up^ CID 12597 from PubChem
  3. Jump up^ Bersot J.C. (2011). “Efficiency Increase of Poly (ethylene terephthalate‐co‐isosorbide terephthalate) Synthesis using Bimetallic Catalytic Systems”. Macromol. Chem. Phys212 (19): 2114–2120. doi:10.1002/macp.201100146.
Isosorbide
Isosorbide.svg
Names
Other names
D-Isosorbide; 1,4:3,6-Dianhydro-D-sorbitol; 1,4-Dianhydrosorbitol
Identifiers
3D model (JSmol)
ChemSpider
ECHA InfoCard 100.010.449
KEGG
PubChem CID
UNII
Properties
C6H10O4
Molar mass 146.14 g·mol−1
Appearance Highly hygroscopic white flakes
Density 1.30 at 25 °C
Melting point 62.5 to 63 °C (144.5 to 145.4 °F; 335.6 to 336.1 K)
Boiling point 160 °C (320 °F; 433 K) at 10 mmHg
in water (>850 g/L), alcoholsand ketones
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

From the net

STR1

Image result for ISOSORBIDE SYNTHESIS

 

 

1H Nuclear magnetic resonance (NMR) spectra of PTMG, isosorbide, HDI, and polyurethane.HDI: hexamethylene diisocyanate; PTMG: poly(tetramethylene glycol).

1H Nuclear magnetic resonance (NMR) spectra of PTMG, isosorbide, HDI, and polyurethane.HDI: hexamethylene diisocyanate; PTMG: poly(tetramethylene glycol).

 

Image result for ISOSORBIDE SYNTHESIS

REF

http://www.rsc.org/suppdata/gc/c4/c4gc01822b/c4gc01822b1.pdf

Synthesis of five- and six-membered heterocycles by dimethyl carbonate with catalytic amount of nitrogen bicyclic bases

http://pubs.rsc.org/en/content/articlelanding/2015/gc/c4gc01822b#!divAbstract

F. Aricò, a,*S. Evaristoa and P. Tundoa,*

Catalytic amount of a nitrogen bicyclic base, i.e., DABCO, DBU and TBD is effective for the one-pot synthesis of heterocycles from 1,4-, 1,5-diols and 1,4-bifunctional compounds via dimethyl carbonate chemistry under neat conditions. Nitrogen bicyclic bases, that previously showed to enhance the reactivity of DMC in methoxycarbonylation reaction by BAc2 mechanism, are herein used for the first time as efficient catalysts for cyclization reaction encompassing both BAc2 and BAl2 pathways. This synthetic procedure was also applied to a large scale synthesis of cyclic sugars isosorbide and isomannide starting from D-sorbitol and D-mannitol, respectively. The resulting anhydro sugar alcohols were obtained as pure crystalline compounds that did not require any further purification or crystallization.

Image result for ISOSORBIDE SYNTHESIS

Larger scale synthesis of isosorbide: In a round bottom flask equipped with a reflux condenser, D-sorbitol (0.05 mol, 1.00 mol. eq.), DMC (0.44 mol, 8.00 mol. eq.), DBU (2.70 mmol, 0.05 mol. eq.) and MeOH (20.00 mL) were heated at reflux while stirring. The progress of the reaction was monitored by NMR. After 48 hours the reaction was stopped, cooled at room temperature and the mixture was filtered over Gooch n°4. Finally, DMC was evaporated under vacuum and the product was obtained as pure in 98% yield (7.90 g, 0.05 mol). Characterization data were consistent with those obtained for the commercially available compound.

STR1

 

Image result for ISOSORBIDE SYNTHESIS

File:Isosorbide dinitrate synthesis.png

 

STR1

 

Image result for ISOSORBIDE SYNTHESIS


Filed under: PROCESS Tagged: isosorbide

Amantadine Hydrochloride, アダマンタン-1-アミン

$
0
0

Amantadine.svg

ChemSpider 2D Image | Amantadine | C10H17N

Amantadine

  • Molecular Formula C10H17N
  • Average mass 151.249 Da
[768-94-5]
1-ADAMANTAMINE
1-adamantanamine; 1-adamantylamine; 1-aminoadamantane; Amantidine; Aminoadamantane
1-Adamantylamine
1-Aminotricyclo(3.3.1.1(sup 3,7))decane
2204333 [Beilstein]
31377-23-8 [RN]
40933-03-7 [RN]
4-pyridinecarboxylic acid, compd. with tricyclo[3.3.1.13,7]decan-1-amine (1:1)
Journal of the American Chemical Society, 91, p. 6457, 1969 DOI: 10.1021/ja01051a047
Synthesis, p. 457, 1976
Amantadine Hydrochloride - API

AMANTADINE HYDROCHLORIDE

  • Molecular FormulaC10H18ClN
  • Average mass187.710 Da
CAS 665-66-7
SPECTROSCOPY BASE
13 C NMR
RAMAN
MASS
Image result for Amantadine NMR
1H NMR
IR

Amantadine (trade name Symmetrel, by Endo Pharmaceuticals) is a drug that has U.S. Food and Drug Administration approval for use both as an antiviral and an antiparkinsonian drug. It is the organic compound 1-adamantylamine or 1-aminoadamantane, meaning it consists of an adamantane backbone that has an amino group substituted at one of the four methyne positions. Rimantadineis a closely related derivative of adamantane with similar biological properties.

Apart from medical uses, this compound is useful as a building block in organic synthesis, allowing the insertion of an adamantyl group.

According to the U.S. Centers for Disease Control and Prevention (CDC) 100% of seasonal H3N2 and 2009 pandemic flu samples tested showed resistance to adamantanes, and amantadine is no longer recommended for treatment of influenza in the United States. Additionally, its effectiveness as an antiparkinsonian drug is undetermined, with a 2003 Cochrane Review concluding that there was insufficient evidence in support of or against its efficacy and safety.[2]

Medical uses

Parkinson’s disease

Amantadine is used to treat Parkinsons disease, as well as parkinsonism syndromes.[3] A 2003 Cochrane review concluded evidence was inadequate to support the use of amantadine for Parkinson’s disease.[2]

An extended release formulation is used to treat dyskinesia, a side effect of levodopa which is taken by people who have Parkinsons.[4]

Influenza

Amantadine is no longer recommended for treatment of influenza A infection. For the 2008/2009 flu season, the CDC found that 100% of seasonal H3N2 and 2009 pandemic flu samples tested have shown resistance to adamantanes.[5] The CDC issued an alert to doctors to prescribe the neuraminidase inhibitors oseltamivir and zanamivir instead of amantadine and rimantadine for treatment of flu.[6][7] A 2014 Cochrane review did not find benefit for the prevention or treatment of influenza A.[8]

Fatigue in multiple sclerosis

Amantadine also seems to have moderate effects on multiple sclerosis (MS) related fatigue.[9]

Adverse effects

Amantadine has been associated with several central nervous system (CNS) side effects, likely due to amantadine’s dopaminergic and adrenergic activity, and to a lesser extent, its activity as an anticholinergic. CNS side effects include nervousness, anxiety, agitation, insomnia, difficulty in concentrating, and exacerbations of pre-existing seizure disorders and psychiatric symptoms in patients with schizophrenia or Parkinson’s disease. The usefulness of amantadine as an anti-parkinsonian drug is somewhat limited by the need to screen patients for a history of seizures and psychiatric symptoms.

Rare cases of severe skin rashes, such as Stevens-Johnson syndrome,[10] and of suicidal ideation have also been reported in patients treated with amantadine.[11][12]

Livedo reticularis is a possible side effect of amantadine use for Parkinson’s disease.[13]

Influenza

The mechanisms for amantadine’s antiviral and antiparkinsonian effects are unrelated. The mechanism of amantadine’s antiviral activity involves interference with the viral protein, M2, a proton channel.[14][15] After entry of the virus into cells via endocytosis, it is localized in acidic vacuoles; the M2 channel functions in transporting protons with the gradient from the vacuolar space into the interior of the virion. Acidification of the interior results in disassociation of ribonucleoproteins, and the initiation of viral replication. Amantadine and rimantadine function in a mechanistically identical fashion in entering the barrel of the tetrameric M2 channel, and blocking pore function (i.e., proton translocation). Resistance to the drug class is a consequence of mutations to the pore-lining residues of the channel, leading to the inability of the sterically bulky adamantane ring that both amantadine and rimantadine share, in entering in their usual way, into the channel.[citation needed]

Influenza B strains possess a structurally distinct M2 channels with channel-facing side chains that fully obstruct the channel vis-a-vis binding of adamantine-class channel inhibitors, while still allowing proton flow and channel function to occur; this constriction in the channels is responsible for the ineffectiveness of this drug and rimantadine towards all circulating Influenza B strains.

Parkinson’s disease

Amantadine is a weak antagonist of the NMDA-type glutamate receptorincreases dopamine release, and blocks dopamine reuptake.[16] Amantadine probably does not inhibit MAO enzyme.[17] Moreover, the mechanism of its antiparkinsonian effect is poorly understood.[citation needed] The drug has many effects in the brain, including release of dopamine and norepinephrine from nerve endings. It appears to be a weak NMDA receptor antagonist[18][19] as well as an anticholinergic, specifically a nicotinic alpha-7 antagonist like the similar pharmaceutical memantine.

In 2004, it was discovered that amantadine and memantine bind to and act as agonists of the σ1 receptor (Ki = 7.44 µM and 2.60 µM, respectively), and that activation of the σ1receptor is involved in the dopaminergic effects of amantadine at therapeutically relevant concentrations.[20] These findings may also extend to the other adamantanes such as adapromine, rimantadine, and bromantane, and could explain the psychostimulant-like effects of this family of compounds.[20]

History

Amantadine was approved by the U.S. Food and Drug Administration in October 1966 as a prophylactic agent against Asian influenza, and eventually received approval for the treatment of influenzavirus A[21][22][23][24] in adults. In 1969, the drug was also discovered by accident upon trying to help reduce symptoms of Parkinson’s disease, drug-induced extrapyramidal syndromes, and akathisia.

In 2017, the U.S. Food and Drug Administration approved the use of amantadine in an extended release formulation developed by Adamas Pharma for the treatment of dyskinesia, an adverse effect of levodopa, that people with Parkinson’s experience.[25]

Veterinary misuse

In 2005, Chinese poultry farmers were reported to have used amantadine to protect birds against avian influenza.[26] In Western countries and according to international livestock regulations, amantadine is approved only for use in humans. Chickens in China have received an estimated 2.6 billion doses of amantadine.[26] Avian flu (H5N1) strains in China and southeast Asia are now resistant to amantadine, although strains circulating elsewhere still seem to be sensitive. If amantadine-resistant strains of the virus spread, the drugs of choice in an avian flu outbreak will probably be restricted to the scarcer and costlier oseltamivir and zanamivir, which work by a different mechanism and are less likely to trigger resistance.

On September 23, 2015, the US Food and Drug Administration announced the recall of Dingo Chip Twists “Chicken in the Middle” dog treats because the product has the potential to be contaminated with amantadine.[27]

Image result for Amantadine SYNTHESIS

Image result for Amantadine SYNTHESIS

Image result for Amantadine SYNTHESIS

PAPER

An Improved Synthesis of Amantadine Hydrochloride

http://pubs.acs.org/doi/10.1021/acs.oprd.7b00242

 Vietnam Military Medical University, No. 160, Phung Hung str., Phuc La ward, Ha Dong district, Hanoi, Vietnam
 School of Chemical Engineering, Hanoi University of Science and Technology, No.1, Dai Co Viet str., Bach Khoa ward, Hai Ba Trung district, Hanoi, Vietnam
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00242
Abstract Image

Amantadine hydrochloride 1 is an antiviral drug used in the prevention and treatment of influenza A infections. It has also been used for alleviating early symptoms of Parkinson’s disease. Several methods for the preparation of 1 have been reported. These procedures started with adamantane 2 using as many as four reaction steps to produce amantadine hydrochloride with overall yields ranging from 45% to 58%. In this article, we describe a two-step procedure for the synthesis of 1from 2 via N-(1-adamantyl)acetamide 4 with an improved overall yield of 67%. The procedure was also optimized to reduce the use of toxic solvents and reagents, rendering it more environment-friendly. The procedure can be considered as suitable for large-scale production of amantadine hydrochloride. The structure of amantadine hydrochloride was confirmed by 1H NMR, 13C NMR, IR, and MS.

Amantadine Hydrochloride (1)

 1. Yield: 232 g (82%). Rf = 0.5 (CHCl3/MeOH/25% aqueous NH3 = 6:1:1).
Purity (GC): 99.22%, tR 10.10 min; mp 360 °C.
1H NMR (CDCl3, 500 MHz): δ 8.28 (br, s, 3H), 2.15 (s, 3H), 2.04 (s, 6H); 1.69 (s, 6H).
13C NMR (CDCl3, 125 MHz): δ 52.95, 40.56, 35.38, 28.97.
IR (KBr): cm–1 3331.73–3185.17 (N–H); 3054.60–2917.82 (C–H); 1363.50 (C–N).
MS: m/z = 151.9 [M + 1]+, 135.0 [M–NH2 – 1]+.
IR spectrum of amantadine hydrochloride (1)
MS spectrum of amantadine hydrochloride
1H-NMR spectrum of amantadine hydrochloride (1) in CDCl3
13C-NMR spectrum of amantadine hydrochloride (1) in CDCl3
Amantadine
Title: Amantadine
CAS Registry Number: 768-94-5
CAS Name: Tricyclo[3.3.1.13,7]decan-1-amine
Additional Names: 1-adamantanamine; 1-aminoadamantane; 1-aminodiamantane (obsolete); 1-aminotricyclo[3.3.1.13,7]decane
Molecular Formula: C10H17N
Molecular Weight: 151.25
Percent Composition: C 79.41%, H 11.33%, N 9.26%
Literature References: NMDA-receptor antagonist; also active vs influenza A virus. Prepn: H. Stetter et al., Ber. 93, 226 (1960); W. Haaf, ibid. 97, 3234 (1964); P. Kovacic, P. D. Roskos, Tetrahedron Lett. 1968, 5833. Antiviral activity: W. L. Davies et al.,Science 144, 862 (1964). GC determn in biological samples and pharmacodynamics: W. E. Bleidner et al., J. Pharmacol. Exp. Ther. 150, 484 (1965). Pharmacology and toxicology: V. G. Vernier et al., Toxicol. Appl. Pharmacol. 15, 642 (1969). Comprehensive description: J. Kirschbaum, Anal. Profiles Drug Subs. 12, 1-36 (1983). Review of use vs influenza A: R. L. Tominack, F. G. Hayden, Infect. Dis. Clin. North Am. 1, 459-478 (1987); of pharmacokinetics: F. Y. Aoki, D. S. Sitar, Clin. Pharmacokinet. 14, 35-51 (1988). Review of NMDA receptor binding and neuroprotective properties: J. Kornhuber et al., J. Neural Transm. 43, Suppl., 91-104 (1994). Series of articles on clinical experience in Parkinson’s disease: ibid. 46, Suppl., 399-421 (1995).
Properties: Crystals by sublimation, mp 160-190° (closed tube) (Stetter). Also reported as mp 180-192° (Haaf). pKa: 10.1. Sparingly sol in water.
Melting point: mp 160-190° (closed tube) (Stetter); mp 180-192° (Haaf)
pKa: pKa: 10.1

Derivative Type: Hydrochloride

CAS Registry Number: 665-66-7
Manufacturers’ Codes: EXP-105-1; NSC-83653
Trademarks: Adekin (Desitin); Lysovir (Alliance); Mantadan (Boehringer, Ing.); Mantadine (Endo); Mantadix (BMS); Symmetrel (Endo); Virofral (Novo)
Molecular Formula: C10H17N.HCl
Molecular Weight: 187.71
Percent Composition: C 63.99%, H 9.67%, N 7.46%, Cl 18.89%
Properties: Crystals from abs ethanol + anhydr ether, mp >360° (dec). Freely sol in water (at least 1:20); sol in alcohol, chloroform. Practically insol in ether. LD50 orally in mice, rats: 700, 1275 mg/kg (Vernier).
Melting point: mp >360° (dec)
Toxicity data: LD50 orally in mice, rats: 700, 1275 mg/kg (Vernier)
Derivative Type: Sulfate
CAS Registry Number: 31377-23-8
Trademarks: PK-Merz (Merz)
Molecular Formula: C10H17N.½H2SO4
Molecular Weight: 200.29
Percent Composition: C 59.97%, H 9.06%, N 6.99%, S 8.00%, O 15.98%
Therap-Cat: Antiviral; antiparkinsonian.
Keywords: Antidyskinetic; Antiparkinsonian; Antiviral.
Amantadine
Amantadine.svg
Amantadine ball-and-stick model.png
Clinical data
Trade names Symmetrel
Synonyms 1-Adamantylamine
AHFS/Drugs.com Monograph
MedlinePlus a682064
Pregnancy
category
  • AU: B3
  • US: C (Risk not ruled out)
Routes of
administration
Oral
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability 86–90%[1]
Protein binding 67%[1]
Metabolism Minimal (mostly to acetyl metabolites)[1]
Biological half-life 10–31 hours[1]
Excretion Urine[1]
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.011.092
Chemical and physical data
Formula C10H17N
Molar mass 151.249 g/mol
3D model (JSmol)

References

  1. Jump up to:a b c d e “SYMMETREL® (amantadine hydrochloride)” (PDF). TGA eBusiness Services. NOVARTIS Pharmaceuticals Australia Pty Limited. 29 June 2011. Retrieved 24 February2014.
  2. Jump up to:a b Crosby, Niall J; Deane, Katherine; Clarke, Carl E (2003). Clarke, Carl E, ed. “Amantadine in Parkinson’s disease”. Cochrane Database of Systematic Reviewsdoi:10.1002/14651858.CD003468.
  3. Jump up^ “Amantadine – FDA prescribing information,”Drugs.com. Retrieved 2017-08-28.
  4. Jump up^ “Amantadine extended release capsules” (PDF). FDA. August 2017. For label updates, see FDA index page for NDA 208944
  5. Jump up^ CDC weekly influenza report – week 35, cdc.gov
  6. Jump up^ “CDC Recommends against the Use of Amantadine and Rimantadine for the Treatment or Prophylaxis of Influenza in the United States during the 2005–06 Influenza Season”CDC Health AlertCenters for Disease Control and Prevention. 2006-01-14. Archived from the original on 3 May 2008. Retrieved 2008-05-20.
  7. Jump up^ Deyde, Varough M.; Xu, Xiyan; Bright, Rick A.; Shaw, Michael; Smith, Catherine B.; Zhang, Ye; Shu, Yuelong; Gubareva, Larisa V.; Cox, Nancy J.; Klimov, Alexander I. (2007). “Surveillance of Resistance to Adamantanes among Influenza A(H3N2) and A(H1N1) Viruses Isolated Worldwide”. Journal of Infectious Diseases196 (2): 249–257. PMID 17570112doi:10.1086/518936.
  8. Jump up^ Alves Galvão, MG; Rocha Crispino Santos, MA; Alves da Cunha, AJ (21 November 2014). “Amantadine and rimantadine for influenza A in children and the elderly.”. The Cochrane database of systematic reviews11: CD002745. PMID 25415374doi:10.1002/14651858.CD002745.pub4.
  9. Jump up^ Braley, TJ; Chervin, RD (Aug 2010). “Fatigue in multiple sclerosis: mechanisms, evaluation, and treatment.”Sleep33 (8): 1061–7. PMC 2910465Freely accessiblePMID 20815187.
  10. Jump up^ Singhal, KC; Rahman, SZ (2002). “Stevens Johnson Syndrome Induced by Amantadine”. Rational Drug Bulletin12 (1): 6.
  11. Jump up^ “Symmetrel (Amantadine) Prescribing Information” (PDF). Endo Pharmaceuticals. May 2003. Retrieved 2007-08-02.
  12. Jump up^ Cook, PE; Dermer, SW; McGurk, T (1986). “Fatal overdose with amantadine”. Canadian Journal of Psychiatry31 (8): 757–8. PMID 3791133.
  13. Jump up^ Vollum, DI; Parkes, JD; Doyle, D (June 1971). “Livedo reticularis during amantadine treatment”Br Med J2 (5762): 627–8. PMC 1796527Freely accessiblePMID 5580722doi:10.1136/bmj.2.5762.627.
  14. Jump up^ Wang C, Takeuchi K, Pinto LH, Lamb RA (1993). “Ion channel activity of influenza A virus M2 protein: characterization of the amantadine block”Journal of Virology67 (9): 5585–94. PMC 237962Freely accessiblePMID 7688826.
  15. Jump up^ Jing X, Ma C, Ohigashi Y, et al. (2008). “Functional studies indicate amantadine binds to the pore of the influenza A virus M2 proton-selective ion channel”Proc. Natl. Acad. Sci. U.S.A105 (31): 10967–72. PMC 2492755Freely accessiblePMID 18669647doi:10.1073/pnas.0804958105.
  16. Jump up^ Jasek, W, ed. (2007). Austria-Codex (in German) (62nd ed.). Vienna: Österreichischer Apothekerverlag. p. 3962. ISBN 978-3-85200-181-4.
  17. Jump up^ Strömberg, U.; Svensson, T. H. (November 1971). “Further Studies on the Mode of Action of Amantadine”wiley.comActa Pharmacologica et Toxicologica, Nordic Pharmacological Society. 30 (3–4): 161–171. doi:10.1111/j.1600-0773.1971.tb00646.x.
  18. Jump up^ Kornhuber, J; Bormann, J; Hübers, M; Rusche, K; Riederer, P (1991). “Effects of the 1-amino-adamantanes at the MK-801-binding site of the NMDA-receptor-gated ion channel: a human postmortem brain study”. Eur. J. Pharmacol. Mol. Pharmacol. Sect206: 297–300. doi:10.1016/0922-4106(91)90113-v.
  19. Jump up^ Blanpied, TA; Clarke, RJ; Johnson, JW (2005). “Amantadine inhibits NMDA receptors by accelerating channel closure during channel block”. Journal of Neuroscience25 (13): 3312–22. PMID 15800186doi:10.1523/JNEUROSCI.4262-04.2005.
  20. Jump up to:a b Peeters, Magali; Romieu, Pascal; Maurice, Tangui; Su, Tsung-Ping; Maloteaux, Jean-Marie; Hermans, Emmanuel (2004). “Involvement of the sigma1 receptor in the modulation of dopaminergic transmission by amantadine”. European Journal of Neuroscience19 (8): 2212–2220. ISSN 0953-816XPMID 15090047doi:10.1111/j.0953-816X.2004.03297.x.
  21. Jump up^ Hounshell, David A.; Kenly Smith, John (1988). Science and Corporate Strategy: Du Pont R&D, 1902–1980. Cambridge University Press. p. 469.
  22. Jump up^ “Sales of flu drug by du Pont unit a ‘disappointment'”The New York Times. Wilmington, Delaware. October 5, 1982. Retrieved May 19, 2008.
  23. Jump up^ Maugh, T. (1979). “Panel urges wide use of antiviral drug”. Science206 (4422): 1058–60. PMID 386515doi:10.1126/science.386515.
  24. Jump up^ Maugh, T. H. (1976). “Amantadine: an Alternative for Prevention of Influenza”. Science192 (4235): 130–1. PMID 17792438doi:10.1126/science.192.4235.130.
  25. Jump up^ Bastings, Eric. “NDA 208944 Approval Letter” (PDF).
  26. Jump up to:a b Sipress, Alan (2005-06-18). “Bird Flu Drug Rendered Useless”Washington Post. pp. A01. Retrieved 2007-08-02.
  27. Jump up^ “Enforcement Report – Week of September 23, 2015”FDA.gov. US Food and Drug Administration, US Department of Health & Human Services.

/////////////


Filed under: Uncategorized Tagged: Amantadine Hydrochloride, アダマンタン-1-アミン

ESCITALOPRAM, S-(+)-Citalopram, эсциталопрам , إيسكيتالوبرام , 艾司西酞普兰 ,

$
0
0
ChemSpider 2D Image | Escitalopram | C20H21FN2OImage result for ESCITALOPRAM
Escitalopram
(+)-Citalopram
(1S)-1-[3-(Dimethylamino)propyl]-1-(4-fluorophenyl)-1,3-dihydro-2-benzofuran-5-carbonitrile [ACD/IUPAC Name]
(S)-citalopram
128196-01-0 [RN]
5-Isobenzofurancarbonitrile, 1-[3-(dimethylamino)propyl]-1-(4-fluorophenyl)-1,3-dihydro-, (1S)- [ACD/Index Name]
  • Molecular FormulaC20H21FN2O
  • Average mass324.392 Da
  • S-(+)-Citalopram
    эсциталопрам [Russian] [INN]
    إيسكيتالوبرام [Arabic] [INN]
    艾司西酞普兰 [Chinese] [INN]

Image result for ESCITALOPRAM

Lexapro® (escitalopram oxalate) is an orally administered selective serotonin reuptake inhibitor (SSRI). Escitalopram is the pure Senantiomer (single isomer) of the racemic bicyclic phthalane derivative citalopram. Escitalopram oxalate is designated S-(+)-1-[3(dimethyl-amino)propyl]-1-(p-fluorophenyl)-5-phthalancarbonitrile oxalate with the following structural formula:

 

Lexapro® (escitalopram oxalate) Structural Formual Illustration

The molecular formula is C20H21FN2O • C2H2O4 and the molecular weight is 414.40.

Escitalopram oxalate occurs as a fine, white to slightly-yellow powder and is freely soluble in methanol and dimethyl sulfoxide (DMSO), soluble in isotonic saline solution, sparingly soluble in water and ethanol, slightly soluble in ethyl acetate, and insoluble in heptane.

Lexapro (escitalopram oxalate) is available as tablets or as an oral solution.

Lexapro tablets are film-coated, round tablets containing escitalopram oxalate in strengths equivalent to 5 mg, 10 mg, and 20 mg escitalopram base. The 10 and 20 mg tablets are scored. The tablets also contain the following inactive ingredients: talc, croscarmellose sodium, microcrystalline cellulose/colloidal silicon dioxide, and magnesium stearate. The film coating contains hypromellose, titanium dioxide, and polyethylene glycol.

Lexapro oral solution contains escitalopram oxalate equivalent to 1 mg/mL escitalopram base. It also contains the following inactive ingredients: sorbitol, purified water, citric acid, sodium citrate, malic acid, glycerin, propylene glycol, methylparaben, propylparaben, and natural peppermint flavor.

Escitalopram, also known by the brand names Lexapro and Cipralex among others, is an antidepressant of the selective serotonin reuptake inhibitor (SSRI) class. It is approved by the U.S. Food and Drug Administration (FDA) for the treatment of adults and children over 12 years of age with major depressive disorder (MDD) or generalized anxiety disorder (GAD). Escitalopram is the (S)-stereoisomer(Left-enantiomer) of the earlier Lundbeck drug citalopram, hence the name escitalopram. Whether escitalopram exhibits superior therapeutic properties to citalopram or merely represents an example of “evergreening” is controversial.[2]

Medical uses

Escitalopram has FDA approval for the treatment of major depressive disorder in adolescents and adults, and generalized anxiety disorder in adults.[3] In European countries and Australia, it is approved for depression (MDD) and certain anxiety disorders: general anxiety disorder (GAD), social anxiety disorder (SAD), obsessive-compulsive disorder (OCD), and panic disorder with or without agoraphobia.

Depression

Escitalopram was approved by regulatory authorities for the treatment of major depressive disorder on the basis of four placebo controlled, double-blind trials, three of which demonstrated a statistical superiority over placebo.[4]

Controversy exists regarding the effectiveness of escitalopram compared to its predecessor citalopram. The importance of this issue follows from the greater cost of escitalopram relative to the generic mixture of isomers citalopram prior to the expiration of the escitalopram patent in 2012, which led to charges of evergreening. Accordingly, this issue has been examined in at least 10 different systematic reviews and meta analyses. The most recent of these have concluded (with caveats in some cases) that escitalopram is modestly superior to citalopram in efficacy and tolerability.[5][6][7][8]

In contrast to these findings, a 2011 review concluded that all second-generation antidepressants are equally effective,[9] and treatment guidelines issued by the National Institute of Health and Clinical Excellence and by the American Psychiatric Association generally reflect this viewpoint.[10][11]

Anxiety disorder

Escitalopram appears to be effective in treating general anxiety disorder, with relapse on escitalopram (20%) less than placebo (50%).[12]

Other

Escitalopram as well as other SSRIs are effective in reducing the symptoms of premenstrual syndrome, whether taken in the luteal phase only or continuously.[13] There is no good data available for escitalopram for seasonal affective disorder as of 2011.[14] SSRIs do not appear to be useful for preventing tension headaches or migraines.[15][16]

Adverse effects

Escitalopram, like other SSRIs, has been shown to affect sexual functions causing side effects such as decreased libidodelayed ejaculation, genital anesthesia,[17] and anorgasmia.[18][19]

An analysis conducted by the FDA found a statistically insignificant 1.5 to 2.4-fold (depending on the statistical technique used) increase of suicidality among the adults treated with escitalopram for psychiatric indications.[20][21][22] The authors of a related study note the general problem with statistical approaches: due to the rarity of suicidal events in clinical trials, it is hard to draw firm conclusions with a sample smaller than two million patients.[23]

Escitalopram is not associated with significant weight gain. For example, 0.6 kg mean weight change after 6 months of treatment with escitalopram for depression was insignificant and similar to that with placebo (0.2 kg).[24] 1.4–1.8 kg mean weight gain was reported in 8-month trials of escitalopram for depression,[25] and generalized anxiety disorder.[26] A 52-week trial of escitalopram for the long-term treatment of depression in elderly also found insignificant 0.6 kg mean weight gain.[27] Escitalopram may help reduce weight in those treated for binge eating associated obesity.[28]

Citalopram and escitalopram are associated with dose-dependent QT interval prolongation[29] and should not be used in those with congenital long QT syndrome or known pre-existing QT interval prolongation, or in combination with other medicines that prolong the QT interval. ECG measurements should be considered for patients with cardiac disease, and electrolyte disturbances should be corrected before starting treatment. In December 2011, the UK implemented new restrictions on the maximum daily doses.[30][31] The U.S. Food and Drug Administration and Health Canada did not similarly order restrictions on escitalopram dosage, only on its predecessor citalopram.[32]

Escitalopram should be taken with caution when using Saint John’s wort.[33] Exposure to escitalopram is increased moderately, by about 50%, when it is taken with omeprazole. The authors of this study suggested that this increase is unlikely to be of clinical concern.[34] Caution should be used when taking cough medicine containing dextromethorphan (DXM) as serotonin syndrome, liver damage, and other negative side effects have been reported.

Discontinuation symptoms

Escitalopram discontinuation, particularly abruptly, may cause certain withdrawal symptoms such as “electric shock” sensations[35] (also known as “brain shivers” or “brain zaps”), dizziness, acute depressions and irritability, as well as heightened senses of akathisia.[36]

Pregnancy

There is a tentative association of SSRI use during pregnancy with heart problems in the baby.[37] Their use during pregnancy should thus be balanced against that of depression.[37]

Overdose

Excessive doses of escitalopram usually cause relatively minor untoward effects such as agitation and tachycardia. However, dyskinesiahypertonia, and clonus may occur in some cases. Plasma escitalopram concentrations are usually in a range of 20–80 μg/L in therapeutic situations and may reach 80–200 μg/L in the elderly, patients with hepatic dysfunction, those who are poor CYP2C19 metabolizers or following acute overdose. Monitoring of the drug in plasma or serum is generally accomplished using chromatographic methods. Chiral techniques are available to distinguish escitalopram from its racemate, citalopram.[38][39][40] Escitalopram seems to be less dangerous than citalopram in overdose and comparable to other SSRIs.[41]

Pharmacology

Mechanism of action

Binding profile[42]
Receptor Ki (nM)
SERT 2.5
NET 6,514
5-HT2C 2,531
α1 3,870
M1 1,242
H1 1,973

Escitalopram increases intrasynaptic levels of the neurotransmitter serotonin by blocking the reuptake of the neurotransmitter into the presynaptic neuron. Of the SSRIs currently on the market, escitalopram has the highest selectivity for the serotonin transporter (SERT) compared to the norepinephrine transporter (NET), making the side-effect profile relatively mild in comparison to less-selective SSRIs.[43] The opposite enantiomer, (R)-citalopram, counteracts to a certain degree the serotonin-enhancing action of escitalopram.[citation needed] As a result, escitalopram has been claimed to be a more potent antidepressant than the racemic mixture, citalopram, of the two enantiomers. In order to explain this phenomenon, researchers from Lundbeck proposed that escitalopram enhances its own binding via an additional interaction with another allosteric site on the transporter.[44] Further research by the same group showed that (R)-citalopram also enhances binding of escitalopram,[45] and therefore the allosteric interaction cannot explain the observed counteracting effect. In the most recent paper, however, the same authors again reversed their findings and reported that (R)-citalopram decreases binding of escitalopram to the transporter.[46] Although allosteric binding of escitalopram to the serotonin transporter is of unquestionable research interest, its clinical relevance is unclear since the binding of escitalopram to the allosteric site is at least 1000 times weaker than to the primary binding site.

Escitalopram is a substrate of P-glycoprotein and hence P-glycoprotein inhibitors such as verapamil and quinidine may improve its blood-brain penetrability.[47] In a preclinical study in rats combining escitalopram with a P-glycoprotein inhibitor enhanced its antidepressant-like effects.[47]

Interactions

Escitalopram, similarly to other SSRIs (with the exception of fluvoxamine), inhibits CYP2D6 and hence may increase plasma levels of a number of CYP2D6 substrates such as aripiprazolerisperidonetramadolcodeine, etc. As much of the effect of codeine is attributable to its conversion (10%) to morphine its effectiveness will be reduced by this inhibition, not enhanced.[48] As escitalopram is only a weak inhibitor of CYP2D6, analgesia from tramadol may not be affected.[49] Escitalopram can also prolong the QT interval and hence it is not recommended in patients that are concurrently on other medications that have the ability to prolong the QT interval. Being a SSRI, escitalopram should not be given concurrently with MAOIs or other serotonergic medications.[43]

History

Cipralex brand escitalopram 10mg package and tablet sheet

Escitalopram was developed in close cooperation between Lundbeck and Forest Laboratories. Its development was initiated in the summer of 1997, and the resulting new drug application was submitted to the U.S. FDA in March 2001. The short time (3.5 years) it took to develop escitalopram can be attributed to the previous extensive experience of Lundbeck and Forest with citalopram, which has similar pharmacology.[50] The FDA issued the approval of escitalopram for major depression in August 2002 and for generalized anxiety disorder in December 2003. On May 23, 2006, the FDA approved a generic version of escitalopram by Teva.[51] On July 14 of that year, however, the U.S. District Court of Delaware decided in favor of Lundbeck regarding the patent infringement dispute and ruled the patent on escitalopram valid.[52]

In 2006 Forest Laboratories was granted an 828-day (2 years and 3 months) extension on its US patent for escitalopram.[53] This pushed the patent expiration date from December 7, 2009 to September 14, 2011. Together with the 6-month pediatric exclusivity, the final expiration date was March 14, 2012.

Society and culture

Allegations of illegal marketing

In 2004, two separate civil suits alleging illegal marketing of citalopram and escitalopram for use by children and teenagers by Forest were initiated by two whistleblowers, one by a practicing physician named Joseph Piacentile, and the other by a Forest salesman named Christopher Gobble.[54] In February 2009, these two suits received support from the US Attorney for Massachusetts and were combined into one. Eleven states and the District of Columbia have also filed notices of intention to intervene as plaintiffs in the action. The suits allege that Forest illegally engaged in off-label promoting of Lexapro for use in children, that the company hid the results of a study showing lack of effectiveness in children, and that the company paid kickbacks to doctors to induce them to prescribe Lexapro to children. It was also alleged that the company conducted so-called “seeding studies” that were, in reality, marketing efforts to promote the drug’s use by doctors.[55][56] Forest responded to these allegations that it “is committed to adhering to the highest ethical and legal standards, and off-label promotion and improper payments to medical providers have consistently been against Forest policy.”[57] In 2010 Forest Pharmaceuticals Inc., agreed to pay more than $313 million to settle the charges over Lexapro and two other drugs, Levothroid and Celexa.[58]

Brand names

Escitalopram is sold under many brand names worldwide such as Cipralex.[1]

Image result for ESCITALOPRAM SYNTHESISImage result for ESCITALOPRAM SYNTHESIS

The Grignard condensation of 5-cyanophthalide (I) with 4-fluorophenylmagnesium bromide (II) in THF gives 1-(4-fluorophenyl)-1-hydroxy-1,3-dihydroisobenzofuran-5-carbonitrile bromomagnesium salt (III), which slowly rearranges to the benzophenone (IV). A new Grignard condensation of (IV) with 3-(dimethylamino)propylmagnesium chloride (V) in THF affords the expected bis(magnesium) salt (VI), which is hydrolyzed with acetic acid to provide the diol (VII) as a racemic mixture. Selective esterification of the primary alcohol of (VII) with (+)-3,3,3-trifluoro-2-methoxy-2-phenylacetyl chloride (VIII) gives the monoester (IX) as a mixture of diastereomers. This mixture is separated by HPLC and the desired diastereomer (X) is treated with potassium tert-butoxide in toluene.

A new method for the preparation of citalopram has been developed: The chlorination of 1-oxo-1,3-dihydroisobenzofuran-5-carboxylic acid (I) with refluxing SOCl2 gives the acyl chloride (II), which is condensed with 2-amino-2-methyl-1-propanol (III) in THF yielding the corresponding amide (IV). The cyclization of (IV) by means of SOCl2 affords the oxazoline (V), which is treated with 4-fluorophenylmagnesium bromide (VI) in THF giving the benzophenone (VII). This compound (VII), without isolation, is treated with 3-(dimethylamino)propylmagnesium chloride (VIII) in the same solvent, providing the cabinol (IX), which is cyclized by means of methanesulfonyl chloride and Et3N in CH2Cl2 yielding the isobenzofuran (X). Finally, this compound is treated with POCl3 in refluxing pyridine to generate the 5-cyano substituent of citalopram.

The chlorination of 1-oxo-1,3-dihydroisobenzofuran-5-carboxylic acid (XII) with refluxing SOCl2 gives the acyl chloride (XIII), which is condensed with 2-amino-2-methyl-1-propanol (XIV) in THF to yield the corresponding amide (XV). The cyclization of (XV) by means of SOCl2 affords the oxazoline (XVI), which is treated with 4-fluorophenylmagnesium bromide (XVII) in THF to give the benzophenone (XVIII). This compound (XVIII), without isolation, is treated with 3-(dimethylamino)propylmagnesium chloride (XIX) in the same solvent to provide the carbinol (XX), which is submitted to optical resolution with (+)- or (-)-tartaric acid, or (+)- or (-)-camphor-10-sulfonic acid (CSA) to give the desired (S)-enantiomer (XXI). Cyclization of (XXI) by means of methanesulfonyl chloride and TEA in dichloromethane yields the chiral isobenzofuran (XXII), which is finally treated with POCl3 in refluxing pyridine.

The Grignard condensation of 5-cyanophthalide (I) with 4-fluorophenylmagnesium bromide (II) in THF gives 1-(4-fluorophenyl)-1-hydroxy-1,3-dihydroisobenzofuran-5-carbonitrile bromomagnesium salt (III), which slowly rearranges to the benzophenone (IV). A new Grignard condensation of (IV) with 3-(dimethylamino)propylmagnesium chloride (V) in THF affords the expected bis(magnesium) salt (VI), which is hydrolyzed with acetic acid to provide the diol (VII) as a racemic mixture. Selective esterification of the primary alcohol of (VII) with (+)-3,3,3-trifluoro-2-methoxy-2-phenylacetyl chloride (VIII) gives the monoester (IX) as a mixture of diastereomers. This mixture is separated by HPLC and the desired diastereomer (X) is treated with potassium tert-butoxide in toluene

The Grignard condensation of 5-cyanophthalide (I) with 4-fluorophenylmagnesium bromide (II) in THF gives 1-(4-fluorophenyl)-1-hydroxy-1,3-dihydroisobenzofuran-5-carbonitrile bromomagnesium salt (III), which slowly rearranges to the benzophenone (IV). A new Grignard condensation of (IV) with 3-(dimethylamino)propylmagnesium chloride (V) in THF affords the expected bis(magnesium) salt (VI), which is hydrolyzed with acetic acid to provide the diol (VII) as a racemic mixture. Selective esterification of the primary alcohol of (VII) with (+)-3,3,3-trifluoro-2-methoxy-2-phenylacetyl chloride (VIII) gives the monoester (IX) as a mixture of diastereomers. This mixture is separated by HPLC and the desired diastereomer (X) is treated with potassium tert-butoxide in toluene.

Racemic 5-bromo-1-[3-(dimethylamino)propyl]-1-(4-fluorophenyl)-1,3-dihydroisobenzofuran (I) is submitted to optical resolution by chiral chromatography to give the corresponding (S)-isomer (II), which is treated with Zn(CN)2 and Pd(PPh3)4 to afford the target Escitalopram.

The esterification of racemic 1-[4-bromo-2-(hydroxymethyl)phenyl]-4-(dimethylamino)-1-(4-fluorophenyl)-1-butanol (I) with (S)-2-(6-methoxynaphth-2-yl)propionyl chloride (II) by means of TEA and DMAP in THF gives the corresponding ester (III) as a diastereomeric mixture that is separated by chiral chromatography over Daicel AD, the desired diastereomer (IV) is easily isolated. Finally, this ester is hydrolyzed and simultaneously cyclized by means of NaH in DMF to provide the target intermediate (V). Other acyl chlorides such as (S)-2-(4-isobutylphenyl)propionyl chloride, (S)-O-acetylmandeloyl chloride, (S)-benzyloxycarbonylprolyl chloride, (S)-2-phenylbutyryl chloride, (S)-2-methoxy-2-phenylacetyl chloride or (S)-N-acetylalanine can also be used in the preceding sequence.

Citalopram
Title: Citalopram
CAS Registry Number: 59729-33-8
CAS Name: 1-[3-(Dimethylamino)propyl]-1-(4-fluorophenyl)-1,3-dihydro-5-isobenzofurancarbonitrile
Additional Names: 1-[3-(dimethylamino)propyl]-1-(4-fluorophenyl)-5-phthalancarbonitrile; nitalapram
Manufacturers’ Codes: Lu-10-171
Molecular Formula: C20H21FN2O
Molecular Weight: 324.39
Percent Composition: C 74.05%, H 6.53%, F 5.86%, N 8.64%, O 4.93%
Literature References: Selective serotonin reuptake inhibitor (SSRI). Prepn: K. P. Boegesoe, A. S. Toft, DE 2657013eidem, US4136193 (1977, 1979 both to Kefalas); A. J. Bigler et al., Eur. J. Med. Chem. – Chim. Ther. 12, 289 (1977). Prepn of enantiomers: K. P. Boegesoe, J. Perregaard, EP 347066eidemUS 4943590, reissued as US RE 34712 (1989, 1990, 1994 all to Lundbeck). Pharmacology: A. V. Christensen et al., Eur. J. Pharmacol. 41, 153 (1977). HPLC determn in plasma and urine: E. Oyehaug et al.,J. Chromatogr. 308, 199 (1984). Comparative biotransformation of enantiomers: L. L. Von Moltke et al., Drug Metab. Dispos. 29, 1102 (2001). Review of clinical pharmacokinetics: K. Brosen, C. A. Naranjo, Eur. Neuropsychopharmacol. 11, 275-283 (2001). Review of clinical experience in depression: M. B. Keller, J. Clin. Psychiatry 61, 896-908 (2000). Clinical trial of S-form in depression: W. J. Burke et al, ibid63, 331 (2002).
Properties: bp0.03 175-181°.
Boiling point: bp0.03 175-181°
Derivative Type: Hydrobromide
CAS Registry Number: 59729-32-7
Trademarks: Celexa (Forest); Cipramil (Lundbeck); Elopram (Recordati); Seropram (Lundbeck)
Molecular Formula: C20H21FN2O.HBr
Molecular Weight: 405.30
Percent Composition: C 59.27%, H 5.47%, F 4.69%, N 6.91%, O 3.95%, Br 19.71%
Properties: Crystals from isopropanol, mp 182-183°.
Melting point: mp 182-183°
Derivative Type: S-(+)-Form
CAS Registry Number: 128196-01-0
Additional Names: Escitalopram
Properties: [a]D +12.33° (c = 1 in methanol).
Optical Rotation: [a]D +12.33° (c = 1 in methanol)
Derivative Type: Escitalopram oxalate
CAS Registry Number: 219861-08-2
Manufacturers’ Codes: Lu-26-054-0
Trademarks: Cipralex (Lundbeck); Gaudium (Recordati); Lexapro (Forest)
Molecular Formula: C20H21FN2O.C2H2O4
Molecular Weight: 414.43
Percent Composition: C 63.76%, H 5.59%, F 4.58%, N 6.76%, O 19.30%
Properties: Fine white to slightly yellow powder. Crystals from acetone, mp 147-148°. [a]D +12.31° (c = 1 in methanol). Freely sol in methanol, DMSO; sol in isotonic saline; sparingly sol in water, ethanol; slightly sol in ethyl acetate. Insol in heptane.
Melting point: mp 147-148°
Optical Rotation: [a]D +12.31° (c = 1 in methanol)
Therap-Cat: Antidepressant.
Keywords: Antidepressant; Bicyclics; Serotonin Uptake Inhibitor.

References

  1. Jump up to:a b drugs.com Drugs.com international: Escitalopram Page accessed April 25, 2015
  2. Jump up^ NHS pays millions of pounds more than it needs to for drugsThe Independent. Retrieved 05/10/2011.
  3. Jump up^ “Escitalopram Oxalate”. The American Society of Health-System Pharmacists. Retrieved 3 April 2011.
  4. Jump up^ “www.accessdata.fda.gov” (PDF).
  5. Jump up^ Ramsberg J, Asseburg C, Henriksson M (2012). “Effectiveness and cost-effectiveness of antidepressants in primary care: a multiple treatment comparison meta-analysis and cost-effectiveness model”PLoS ONE7 (8): e42003. PMC 3410906Freely accessiblePMID 22876296doi:10.1371/journal.pone.0042003.
  6. Jump up^ Cipriani A, Purgato M, Furukawa TA, Trespidi C, Imperadore G, Signoretti A, Churchill R, Watanabe N, Barbui C (2012). “Citalopram versus other anti-depressive agents for depression”Cochrane Database Syst Rev7: CD006534. PMC 4204633Freely accessiblePMID 22786497doi:10.1002/14651858.CD006534.pub2.
  7. Jump up^ Favré P (February 2012). “[Clinical efficacy and achievement of a complete remission in depression: increasing interest in treatment with escitalopram]”. Encephale (in French). 38(1): 86–96. PMID 22381728doi:10.1016/j.encep.2011.11.003.
  8. Jump up^ Sicras-Mainar A, Navarro-Artieda R, Blanca-Tamayo M, Gimeno-de la Fuente V, Salvatella-Pasant J (December 2010). “Comparison of escitalopram vs. citalopram and venlafaxine in the treatment of major depression in Spain: clinical and economic consequences”. Curr Med Res Opin26 (12): 2757–64. PMID 21034375doi:10.1185/03007995.2010.529430.
  9. Jump up^ Gartlehner G, Hansen RA, Morgan LC, Thaler K, Lux L, Van Noord M, Mager U, Thieda P, Gaynes BN, Wilkins T, Strobelberger M, Lloyd S, Reichenpfader U, Lohr KN (Dec 6, 2011). “Comparative benefits and harms of second-generation antidepressants for treating major depressive disorder: an updated meta-analysis.”. Annals of Internal Medicine155 (11): 772–85. PMID 22147715doi:10.7326/0003-4819-155-11-201112060-00009.
  10. Jump up^ “CG90 Depression in adults: full guidance”.
  11. Jump up^ “PsychiatryOnline | APA Practice Guidelines | Practice Guideline for the Treatment of Patients With Major Depressive Disorder, Third Edition”.[permanent dead link]
  12. Jump up^ Bech P, Lönn SL, Overø KF (2010). “Relapse prevention and residual symptoms: a closer analysis of placebo-controlled continuation studies with escitalopram in major depressive disorder, generalized anxiety disorder, social anxiety disorder, and obsessive-compulsive disorder”. J Clin Psychiatry71 (2): 121–9. PMID 19961809doi:10.4088/JCP.08m04749blu.
  13. Jump up^ Marjoribanks J, Brown J, O’Brien PM, Wyatt K (Jun 7, 2013). “Selective serotonin reuptake inhibitors for premenstrual syndrome.”. The Cochrane database of systematic reviews6: CD001396. PMID 23744611doi:10.1002/14651858.CD001396.pub3.
  14. Jump up^ Thaler K, Delivuk M, Chapman A, Gaynes BN, Kaminski A, Gartlehner G (Dec 7, 2011). “Second-generation antidepressants for seasonal affective disorder.”. The Cochrane database of systematic reviews (12): CD008591. PMID 22161433doi:10.1002/14651858.CD008591.pub2.
  15. Jump up^ Banzi, R; Cusi, C; Randazzo, C; Sterzi, R; Tedesco, D; Moja, L (1 May 2015). “Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs) for the prevention of tension-type headache in adults.”. The Cochrane database of systematic reviews5: CD011681. PMID 25931277doi:10.1002/14651858.CD011681.
  16. Jump up^ Moja, PL; Cusi, C; Sterzi, RR; Canepari, C (20 July 2005). “Selective serotonin re-uptake inhibitors (SSRIs) for preventing migraine and tension-type headaches.”. The Cochrane database of systematic reviews (3): CD002919. PMID 16034880doi:10.1002/14651858.CD002919.pub2.
  17. Jump up^ Bolton JM, Sareen J, Reiss JP (2006). “Genital anesthesia persisting six years after sertraline discontinuation”. J Sex Marital Ther32 (4): 327–30. PMID 16709553doi:10.1080/00926230600666410.
  18. Jump up^ Clayton A, Keller A, McGarvey EL (2006). “Burden of phase-specific sexual dysfunction with SSRIs”. Journal of Affective Disorders91 (1): 27–32. PMID 16430968doi:10.1016/j.jad.2005.12.007.
  19. Jump up^ “Lexapro prescribing information” (PDF).
  20. Jump up^ Levenson M, Holland C. “Antidepressants and Suicidality in Adults: Statistical Evaluation. (Presentation at Psychopharmacologic Drugs Advisory Committee; December 13, 2006)”. Retrieved 2007-05-13.
  21. Jump up^ Stone MB, Jones ML (2006-11-17). “Clinical Review: Relationship Between Antidepressant Drugs and Suicidality in Adults” (PDF). Overview for December 13 Meeting of Pharmacological Drugs Advisory Committee (PDAC). FDA. pp. 11–74. Retrieved 2007-09-22.
  22. Jump up^ Levenson M; Holland C (2006-11-17). “Statistical Evaluation of Suicidality in Adults Treated with Antidepressants” (PDF). Overview for December 13 Meeting of Pharmacological Drugs Advisory Committee (PDAC). FDA. pp. 75–140. Retrieved 2007-09-22.
  23. Jump up^ Khan A, Schwartz K (2007). “Suicide risk and symptom reduction in patients assigned to placebo in duloxetine and escitalopram clinical trials: analysis of the FDA summary basis of approval reports”. Ann Clin Psychiatry19 (1): 31–6. PMID 17453659doi:10.1080/10401230601163550.
  24. Jump up^ Baldwin DS, Reines EH, Guiton C, Weiller E (2007). “Escitalopram therapy for major depression and anxiety disorders”. Ann Pharmacother41 (10): 1583–92. PMID 17848424doi:10.1345/aph.1K089.
  25. Jump up^ Pigott TA, Prakash A, Arnold LM, Aaronson ST, Mallinckrodt CH, Wohlreich MM (2007). “Duloxetine versus escitalopram and placebo: an 8-month, double-blind trial in patients with major depressive disorder”. Curr Med Res Opin23 (6): 1303–18. PMID 17559729doi:10.1185/030079907X188107.
  26. Jump up^ Davidson JR, Bose A, Wang Q (2005). “Safety and efficacy of escitalopram in the long-term treatment of generalized anxiety disorder”. J Clin Psychiatry66 (11): 1441–6. PMID 16420082doi:10.4088/JCP.v66n1115.
  27. Jump up^ Kasper S, Lemming OM, de Swart H (2006). “Escitalopram in the long-term treatment of major depressive disorder in elderly patients”. Neuropsychobiology54 (3): 152–9. PMID 17230032doi:10.1159/000098650.
  28. Jump up^ Guerdjikova AI, McElroy SL, Kotwal R, Welge JA, Nelson E, Lake K, Alessio DD, Keck PE, Hudson JI (January 2008). “High-dose escitalopram in the treatment of binge-eating disorder with obesity: a placebo-controlled monotherapy trial”. Human Psychopharmacology: Clinical and Experimental23 (1): 1–11. PMID 18058852doi:10.1002/hup.899.
  29. Jump up^ Castro VM, Clements CC, Murphy SN, Gainer VS, Fava M, Weilburg JB, Erb JL, Churchill SE, Kohane IS, Iosifescu DV, Smoller JW, Perlis RH (2013). “QT interval and antidepressant use: a cross sectional study of electronic health records”BMJ346: f288. PMC 3558546Freely accessiblePMID 23360890doi:10.1136/bmj.f288.
  30. Jump up^ “Citalopram and escitalopram: QT interval prolongation—new maximum daily dose restrictions (including in elderly patients), contraindications, and warnings”Medicines and Healthcare products Regulatory Agency. December 2011. Retrieved March 5, 2013.
  31. Jump up^ van Gorp F, Whyte IM, Isbister GK (2009). “Clinical and ECG Effects of Escitalopram Overdose” (PDF). Annals of Emergency Medicine54 (3): 404–8. PMID 19556032doi:10.1016/j.annemergmed.2009.04.016.
  32. Jump up^ Hasnain M, Howland RH, Vieweg WV (2013). “Escitalopram and QTc prolongation”J Psychiatry Neurosci38 (4): E11. PMC 3692726Freely accessibledoi:10.1503/jpn.130055.
  33. Jump up^ Karch, Amy (2006). 2006 Lippincott’s Nursing Drug Guide. Philadelphia, Baltimore, New York, London, Buenos Aires, Hong Kong, Sydney, Tokyo: Lippincott Williams & Wilkins. ISBN 1-58255-436-6.
  34. Jump up^ Malling D, Poulsen MN, Søgaard B (2005). “The effect of cimetidine or omeprazole on the pharmacokinetics of escitalopram in healthy subjects”British Journal of Clinical Pharmacology60 (3): 287–290. PMC 1884771Freely accessiblePMID 16120067doi:10.1111/j.1365-2125.2005.02423.x.
  35. Jump up^ Prakash O, Dhar V (2008). “Emergence of electric shock-like sensations on escitalopram discontinuation”. J Clin Psychopharmacol28 (3): 359–60. PMID 18480703doi:10.1097/JCP.0b013e3181727534.
  36. Jump up^ “Lexapro (Escitalopram Oxalate) Drug Information: Warnings and Precautions – Prescribing Information at RxList”. Retrieved 2015-08-09.
  37. Jump up to:a b Gentile, S (1 July 2015). “Early pregnancy exposure to selective serotonin reuptake inhibitors, risks of major structural malformations, and hypothesized teratogenic mechanisms.”. Expert opinion on drug metabolism & toxicology11: 1–13. PMID 26135630doi:10.1517/17425255.2015.1063614.
  38. Jump up^ van Gorp F, Whyte IM, Isbister GK (2009). “Clinical and ECG effects of escitalopram overdose”. Ann Emerg Med54 (3): 404–8. PMID 19556032doi:10.1016/j.annemergmed.2009.04.016.
  39. Jump up^ Haupt D (1996). “Determination of citalopram enantiomers in human plasma by liquid chromatographic separation on a Chiral-AGP column”. J. Chromatogr. B, Biomed. Appl685(2): 299–305. PMID 8953171doi:10.1016/s0378-4347(96)00177-6.
  40. Jump up^ Baselt RC (2008). Disposition of toxic drugs and chemicals in man (8th ed.). Foster City, Ca: Biomedical Publications. pp. 552–553. ISBN 0962652377.
  41. Jump up^ White N, Litovitz T, Clancy C (December 2008). “Suicidal antidepressant overdoses: a comparative analysis by antidepressant type”Journal of Medical Toxicology4 (4): 238–250. PMC 3550116Freely accessiblePMID 19031375doi:10.1007/BF03161207.
  42. Jump up^ Owens, MJ; Knight, DL; Nemeroff, CB (1 September 2001). “Second-generation SSRIs: human monoamine transporter binding profile of escitalopram and R-fluoxetine.”. Biological Psychiatry50 (5): 345–50. PMID 11543737doi:10.1016/s0006-3223(01)01145-3.
  43. Jump up to:a b Brunton L, Chabner B, Knollman B. Goodman and Gilman’s The Pharmacological Basis of Therapeutics, Twelfth Edition. McGraw Hill Professional; 2010.
  44. Jump up^ For an overview of supporting data, see Sánchez C, Bøgesø KP, Ebert B, Reines EH, Braestrup C (2004). “Escitalopram versus citalopram: the surprising role of the R-enantiomer”. Psychopharmacology174 (2): 163–76. PMID 15160261doi:10.1007/s00213-004-1865-z.
  45. Jump up^ Chen F, Larsen MB, Sánchez C, Wiborg O (2005). “The (S)-enantiomer of (R,S)-citalopram, increases inhibitor binding to the human serotonin transporter by an allosteric mechanism. Comparison with other serotonin transporter inhibitors”. European Neuropsychopharmacology15 (2): 193–198. PMID 15695064doi:10.1016/j.euroneuro.2004.08.008.
  46. Jump up^ Mansari ME, Wiborg O, Mnie-Filali O, Benturquia N, Sánchez C, Haddjeri N (2007). “Allosteric modulation of the effect of escitalopram, paroxetine and fluoxetine: in-vitro and in-vivo studies”. The International Journal of Neuropsychopharmacology10 (1): 31–40. PMID 16448580doi:10.1017/S1461145705006462.
  47. Jump up to:a b O’Brien FE, O’Connor RM, Clarke G, Dinan TG, Griffin BT, Cryan JF (October 2013). “P-glycoprotein inhibition increases the brain distribution and antidepressant-like activity of escitalopram in rodents”Neuropsychopharmacology38 (11): 2209–2219. PMC 3773671Freely accessiblePMID 23670590doi:10.1038/npp.2013.120.
  48. Jump up^ Ali Torkamani. “Selective Serotonin Reuptake Inhibitors and CYP2D6”Medscape.com. Retrieved 14 May 2015.
  49. Jump up^ Noehr-Jensen, L; Zwisler, ST; Larsen, F; Sindrup, SH; Damkier, P; Brosen, K (December 2009). “Escitalopram is a weak inhibitor of the CYP2D6-catalyzed O-demethylation of (+)-tramadol but does not reduce the hypoalgesic effect in experimental pain.”. Clinical pharmacology and therapeutics86 (6): 626–33. PMID 19710642doi:10.1038/clpt.2009.154.
  50. Jump up^ “2000 Annual Report. p 28 and 33” (PDF). Lundbeck. 2000. Retrieved 2007-04-07.
  51. Jump up^ Miranda Hitti. “FDA OKs Generic Depression Drug – Generic Version of Lexapro Gets Green Light”. WebMD. Retrieved 2007-10-10.
  52. Jump up^ Marie-Eve Laforte (2006-07-14). “US court upholds Lexapro patent”. FirstWord. Retrieved 2007-10-10.
  53. Jump up^ “Forest Laboratories Receives Patent Term Extension for Lexapro” (Press release). PRNewswire-FirstCall. 2006-03-02. Retrieved 2009-01-19.
  54. Jump up^ “Forest Laboratories: A Tale of Two Whistleblowers” article by Alison Frankel in The American Lawyer February 27, 2009
  55. Jump up^ United States of America v. Forest Laboratories Full text of the federal complaint filed in the US District Court for the district of Massachusetts
  56. Jump up^ “Drug Maker Is Accused of Fraud” article by Barry Meier and Benedict Carey in The New York Times February 25, 2009
  57. Jump up^ “Forest Laboratories, Inc. Provides Statement in Response to Complaint Filed by U.S. Government” Forest press-release. February 26, 2009.
  58. Jump up^ “Drug Maker Forest Pleads Guilty; To Pay More Than $313 Million to Resolve Criminal Charges and False Claims Act Allegations”http://www.justice.gov.

Cited texts

Further reading

External links

Escitalopram
Escitalopram.svg
Escitalopram-from-xtal-3D-balls.png
Clinical data
Pronunciation About this sound pronunciation 
Trade names Cipralex, Lexapro and many others[1]
AHFS/Drugs.com Monograph
MedlinePlus a603005
License data
Pregnancy
category
  • AU: C
  • US: C (Risk not ruled out)
Routes of
administration
Oral
ATC code
Legal status
Legal status
  • AU: S4 (Prescription only)
  • CA℞-only
  • UK: POM (Prescription only)
  • US: ℞-only
  • In general: ℞ (Prescription only)
Pharmacokinetic data
Bioavailability 80%
Protein binding ~56%
Metabolism Liver, specifically the enzymes CYP3A4 and CYP2C19
Biological half-life 27–32 hours
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
ChEBI
ChEMBL
Chemical and physical data
Formula C20H21FN2O
Molar mass 324.392 g/mol
(414.43 as oxalate)
3D model (JSmol)

///////////////////S-(+)-Citalopram, эсциталопрам إيسكيتالوبرام 艾司西酞普兰 , CITALOPRAM

http://shodhganga.inflibnet.ac.in/bitstream/10603/101297/15/15_chapter%206.pdf


Filed under: Uncategorized Tagged: ESCITALOPRAM, 艾司西酞普兰, эсциталопрам, S-(+)-Citalopram, إيسكيتالوبرام

(+)-(S,S)-Reboxetine succinate, Esreboxetine succinate

$
0
0

Image result for (S,S)-Reboxetine succinateimg

Esreboxetine succinate

str1

(2S)-2-[(S)-(2-ethoxyphenoxy)(phenyl)methyl]morpholine butanedioate (1:1)
(2S)-2-[(S)-(2-Ethoxyphenoxy)(phenyl)methyl]morpholine succinate (1:1)
(S,S)-reboxetine succinate
635724-55-9 [RN]
Esreboxetine succinate [USAN]
Morpholine, 2-[(S)-(2-ethoxyphenoxy)phenylmethyl]-, (2S)-, butanedioate (1:1)
Succinic acid – (2S)-2-[(S)-(2-ethoxyphenoxy)(phenyl)methyl]morpholine (1:1)
UNII:XQO13W6OCH

Esreboxetine is a selective norepinephrine reuptake inhibitor which was under development by Pfizer for the treatment of neuropathic pain and fibromyalgia but failed to show significant benefit over currently available medications and was discontinued.[1][2][3][4] It is the (S,S)-(+)-enantiomer of reboxetine and is even more selective in comparison.[1][5]

However, recently it has been shown that esreboxetine could be effective in fibromyalgia patients.[6]

Figure

Reboxetine mesylate (1) and succinate (2).

Image result for (S,S)-Reboxetine succinate

Image result for (S,S)-Reboxetine succinate

CLIP

http://pubs.rsc.org/en/Content/ArticleHtml/2012/GC/c1gc15921f

The synthesis of (±)-reboxetine mesylate,4 the Active Pharmaceutical Ingredient (API) for Edronax™.

Scheme 1 The synthesis of (±)-reboxetine mesylate,4 the Active Pharmaceutical Ingredient (API) for Edronax™.

 

The conversion of (±)-reboxetine mesylate to (S,S)-reboxetine succinate.
Scheme 2 The conversion of (±)-reboxetine mesylate to (S,S)-reboxetine succinate.

 

The Pfizer early resolution route to (S,S)-reboxetine succinate.
Scheme 3 The Pfizer early resolution route to (S,S)-reboxetine succinate.

The Pfizer asymmetric synthesis for (S,S)-reboxetine intended for commercialisation.

Scheme 4 The Pfizer asymmetric synthesis for (S,S)-reboxetine intended for commercialisation.

CLIP

(S,S)-Reboxetine succinate (3) (Figure 1) has been under late-stage development at Pfizer for the medication of neuropathic and fibromyalgia pain.(16)

16.(a) HughesB.McKenzieI.StokerM. J. WO2006/000903, May 1, 2006.

(b) AllenA. J.Hemrick-LueckeS.SumnerC. R.WallaceO. B. WO2005/060949, July 7, 2005.

(c) KelseyD. K. WO2005/021095, Oct 3, 2005.

(d) AllenA. J.KelseyD. K. WO 2005/020976, Oct 3, 2005.

(e) SumnerC. R. WO2005/020975, Oct 3, 2005.

(f) HassanF. WO2004/016272, Feb 26, 2004.

(g) WongE. H. F. WO2004/002463, Jan 8, 2004.

PAPER

Process Development for (S,S)-Reboxetine Succinate via a Sharpless Asymmetric Epoxidation

http://pubs.acs.org/doi/abs/10.1021/op700007g?crel=US_AC_eAdv_Blog

Pfizer Global Research and Development, 2800 Plymouth Road, Ann Arbor, Michigan 48105, U.S.A.
Org. Process Res. Dev.200711 (3), pp 354–358
DOI: 10.1021/op700007g
Publication Date (Web): March 23, 2007
Copyright © 2007 American Chemical Society

Abstract

Abstract Image

Reboxetine mesylate is a selective norepinephrine uptake inhibitor (NRI) currently marketed as the racemate. The (S,S)-enantiomer of reboxetine is being evaluated for the treatment of neuropathic pain and a variety of other indications. (S,S)-Reboxetine has usually been prepared by resolution of the racemate as the (−)-mandelate salt, an inherently inefficient process. A chiral synthesis starting with a Sharpless asymmetric epoxidation of cinnamyl alcohol to yield (R,R)-phenylglycidol was developed. (R,R)-Phenylglycidol was reacted without isolation with 2-ethoxyphenol to give 4, which was isolated by direct crystallization. Key process variables for the asymmetric epoxidation were investigated. Conversion of (R,S)-4 to reboxetine parallels the racemic synthesis with streamlined and optimized processing conditions. (S,S)-Reboxetine free base was converted directly to the succinate salt without isolation as the mesylate salt.

(2S,3S)-Reboxetine Succinate (9).

mp 145.2−147.1 °C (lit. mp 148 °C).8 1H NMR (400.13 MHz, CDCl3) δ 1.41 (t, J = 7.0 Hz, 3H), 2.4 (s, 4H), 2.9−3.06 (m, 2H), 3.15−3.22 (m, 2H), 3.81−3.86 (m, 1H), 4.02−4.09 (m, 3H), 4.17−4.24 (m, 1H), 5.13 (d, J = 4.3 Hz), 6.66−6.90 (m, 4H), 7.26−7.39 (m, 5H). 13C NMR (100.62 MHz, CDCl3) δ 15.08, 31.89, 43.24, 44.84, 64.72, 76.91, 82.91, 113.94, 118.27, 121.1, 127.38, 128.66, 136.94, 149.8, 178.73. LRMS-APCI m/z calcd for C19H23NO3 (M + H)+:  314. Found:  m/z = 314 [M + 1]+. Anal. Calcd for C19H23NO3−C4H6O4:  C, 64.02; H, 6.77; N, 3.25. Found:  C, 63.99; H, 6.77; N, 3.16. [α]32.4D +17.24° (c 0.5, EtOH).

8)Zampieri, M.; Airoldi, A.; Martini, A. WO2003/106441, 12/24/03.

PAPER

Commercial Synthesis of (S,S)-Reboxetine Succinate: A Journey To Find the Cheapest Commercial Chemistry for Manufacture

http://pubs.acs.org/doi/abs/10.1021/op200181f

Chemical Research and Development, Pfizer Inc., Sandwich Laboratories, Sandwich, Kent, CT13 9NJ, United Kingdom
Org. Process Res. Dev.201115 (6), pp 1305–1314
DOI: 10.1021/op200181f
Publication Date (Web): August 18, 2011
Copyright © 2011 American Chemical Society

Abstract

Abstract Image

The development of a synthetic process for (S,S)-reboxetine succinate, a candidate for the treatment of fibromylagia, is disclosed from initial scale-up to deliver material for registrational stability testing through to commercial route evaluation and subsequent nomination. This entailed evaluation of several alternative routes to result in what would have been a commercially attractive process for launch of the compound.

(2S,3S)-2-[α-(2-Ethoxyphenoxy)benzyl]morpholine Succinate Salt (S,S)-Reboxetine Succinate

 (S,S)-reboxetine succinate (897 g, 82%) as a white solid. 1H NMR (400 MHz, d6-DMSO) δ 7.22–7.54 (m, 5H), 6.66–6.96 (m, 4H), 5.27 (d, J = 6.0 Hz, 1H), 4.01 (q, J = 7.1 Hz, 2H), 3.83 (m, 2H), 3.50 (m, 2H), 2.61–2.82 (m, 3H), 2.34 (br s, 4H), 1.33 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, d6-DMSO) δ 174.4, 149.0, 147.3, 137.8, 128.2, 127.3, 120.7, 116.7, 114.4, 80.8, 77.5, 65.9, 64.1, 45.8, 44.1, 39.7, 39.

References[edit]

  1. Jump up to:a b Matilda Bingham; Napier, Susan Jolliffe (2009). Transporters as Targets for Drugs (Topics in Medicinal Chemistry). Berlin: Springer. ISBN 3-540-87911-0.
  2. Jump up^ Rao SG (October 2009). “Current progress in the pharmacological therapy of fibromyalgia”Expert Opinion on Investigational Drugs18 (10): 1479–93. PMID 19732029doi:10.1517/13543780903203771.
  3. Jump up^ “Search of: esreboxetine – List Results – ClinicalTrials.gov”.
  4. Jump up^ “Musculoskeletal Report: Pfizer Stops Work on Esreboxetine for FM”.
  5. Jump up^ Fish, P. V.; MacKenny, M.; Bish, G.; Buxton, T.; Cave, R.; Drouard, D.; Hoople, D.; Jessiman, A.; Miller, D.; Pasquinet, C.; Patel, B.; Reeves, K.; Ryckmans, T.; Skerten, M.; Wakenhut, F. (2009). “Enantioselective synthesis of (R)- and (S)-N-Boc-morpholine-2-carboxylic acids by enzyme-catalyzed kinetic resolution: application to the synthesis of reboxetine analogs”. Tetrahedron Letters50 (4): 389. doi:10.1016/j.tetlet.2008.11.025.
  6. Jump up^ Arnold, L. M., Hirsch, I., Sanders, P., Ellis, A. and Hughes, B. (2012), Safety and efficacy of esreboxetine in patients with fibromyalgia: A fourteen-week, randomized, 

REFERENCES

1: Fujimori I, Yukawa T, Kamei T, Nakada Y, Sakauchi N, Yamada M, Ohba Y, Takiguchi M, Kuno M, Kamo I, Nakagawa H, Hamada T, Igari T, Okuda T, Yamamoto S, Tsukamoto T, Ishichi Y, Ueno H. Design, synthesis and biological evaluation of a novel series of peripheral-selective noradrenaline reuptake inhibitor. Bioorg Med Chem. 2015 Aug 1;23(15):5000-14. doi: 10.1016/j.bmc.2015.05.017. Epub 2015 May 15. PubMed PMID: 26051602.

2: Shen F, Tsuruda PR, Smith JA, Obedencio GP, Martin WJ. Relative contributions of norepinephrine and serotonin transporters to antinociceptive synergy between monoamine reuptake inhibitors and morphine in the rat formalin model. PLoS One. 2013 Sep 30;8(9):e74891. doi: 10.1371/journal.pone.0074891. eCollection 2013. PubMed PMID: 24098676; PubMed Central PMCID: PMC3787017.

3: Arnold LM, Hirsch I, Sanders P, Ellis A, Hughes B. Safety and efficacy of esreboxetine in patients with fibromyalgia: a fourteen-week, randomized, double-blind, placebo-controlled, multicenter clinical trial. Arthritis Rheum. 2012 Jul;64(7):2387-97. doi: 10.1002/art.34390. PubMed PMID: 22275142.

4: Arnold LM, Chatamra K, Hirsch I, Stoker M. Safety and efficacy of esreboxetine in patients with fibromyalgia: An 8-week, multicenter, randomized, double-blind, placebo-controlled study. Clin Ther. 2010 Aug;32(9):1618-32. doi: 10.1016/j.clinthera.2010.08.003. PubMed PMID: 20974319.

5: Klarskov N, Scholfield D, Soma K, Darekar A, Mills I, Lose G. Measurement of urethral closure function in women with stress urinary incontinence. J Urol. 2009 Jun;181(6):2628-33; discussion 2633. doi: 10.1016/j.juro.2009.01.114. Epub 2009 Apr 16. PubMed PMID: 19375093.

Esreboxetine
Esreboxetine.svg
Clinical data
Routes of
administration
Oral
ATC code
  • None
Legal status
Legal status
  • In general: uncontrolled
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
ChemSpider
UNII
KEGG
Chemical and physical data
Formula C19H23NO3
Molar mass 313.391 g/mol
3D model (JSmol)

////////////(+)-(S,S)-Reboxetine, (S,S)-Reboxetine, Reboxetine, Esreboxetine succinate

CCOc1ccccc1O[C@H]([C@@H]2CNCCO2)c3ccccc3.OC(=O)CCC(=O)O


Filed under: Uncategorized Tagged: (+)-(S, Esreboxetine succinate, Reboxetine, S)-Reboxetine, S)-Reboxetine succinate

Hyderabad. India to Host Industrial Organic Chemistry Workshops in February 2018

$
0
0

 

Dr Will Watson, an expert in Chemical Development and related fields, from Scientific Update will be visiting India in February to deliver two important workshops for Industrial Process Chemists:

Chemical Development and Scale Up in the Fine Chemical and Pharmaceutical Industries, February 5th – 7th 2018, Hyderabad, India

Practical Crystallisation & Polymorphism, February 8th & 9th 2018, Hyderabad, India

Discounts are available for groups – please contact sciup@scientificupdate.com for more information.


Filed under: conference Tagged: hyderabad, SCIENTIFIC UPDATE, WILL WATSON

FDA approves CAR-T cell therapy Yescarta (axicabtagene ciloleucel) to treat adults with certain types of large B-cell lymphoma

$
0
0

FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma

Yescarta is the second gene therapy product approval in the U.S.

The U.S. Food and Drug Administration today approved Yescarta (axicabtagene ciloleucel), a cell-based gene therapy, to treat adult patients with certain types of large B-cell lymphoma who have not responded to or who have relapsed after at least two other kinds of treatment. Yescarta, a chimeric antigen receptor (CAR) T cell therapy, is the second gene therapy approved by the FDA and the first for certain types of non-Hodgkin lymphoma (NHL). Continue reading.

/////////FDA, CAR-T cell therapy,  large B-cell lymphoma, fda 2017, Yescarta, axicabtagene ciloleucel,


Filed under: FDA 2017 Tagged: axicabtagene ciloleucel, CAR-T cell therapy, fda, FDA 2017, large B-cell lymphoma, Yescarta

(R)-(–)-Baclofen, Arbaclofen, STX 209, AGI 006

$
0
0

(R)-Baclofen.pngChemSpider 2D Image | Arbaclofen | C10H12ClNO2

(R)-(–)-Baclofen, Arbaclofen, STX 209, AGI 006

Chemical Names: (R)-Baclofen; Arbaclofen; 69308-37-8; (R)-4-Amino-3-(4-chlorophenyl)butanoic acid; (-)-Baclofen; D-Baclofen
Molecular Formula: C10H12ClNO2
Molecular Weight: 213.661 g/mol

 A GAMMA-AMINOBUTYRIC ACID derivative that is a specific agonist of GABA-B RECEPTORS. It is used in the treatment of MUSCLE SPASTICITY, especially that due to SPINAL CORD INJURIES. Its therapeutic effects result from actions at spinal and supraspinal sites, generally the reduction of excitatory transmission.

(R)-4-Amino-3-(4-chlorophenyl)butanoic acid

Benzeneporopanoic acid, (beta-(aminomethyl)-4-chloro-, (betaR)-

Spasticity,  PREREGISTERD, OSMOTICA PHARMA

  • Benzenepropanoic acid, β-(aminomethyl)-4-chloro-, (R)-
  • (βR)-β-(Aminomethyl)-4-chlorobenzenepropanoic acid
  • (-)-Baclofen
  • (R)-(-)-Baclofen
  • (R)-4-Amino-3-(4-chlorophenyl)butanoic acid
  • (R)-4-Amino-3-(4-chlorophenyl)butyric acid
  • (R)-Baclofen
  • AGI 006
  • Arbaclofen
  • D-Baclofen
  • R-(-)-Baclofen
  • STX 209
  • l-Baclofen

Optical Rotatory Power, -1.76 °, Conc: 0.5 g/100mL; Solv: water (7732-18-5); Wavlen: 589.3 nm; Temp: 25 °C, REF …..Paraskar, Abhimanyu S.; Tetrahedron 2006, VOL62(20), PG4907-4916

Melting Point 196-197 °C Solv: isopropanol (67-63-0)

REF…..Paraskar, Abhimanyu S.; Tetrahedron 2006, VOL62(20), PG4907-4916

 

Image result for (R)-(–)-Baclofen

Arbaclofen, or STX209, is the R-enantiomer of baclofen. It is believed to be a selective gamma-amino butyric acid type B receptor agonist, and has been investigated as a treatment for autism spectrum disorder and fragile X syndrome in randomized, double blind, placebo controlled trials. It has also been investigated as a treatment for spasticity due to multiple sclerosis and spinal cord injury. Arbaclofen was investigated as a treatment for gastroesophageal reflux disease (GERD); however, with disappointing results.

AGI-006, a GABA(B) agonist, is currently in phase III clinical trials at Seaside Therapeutics for the treatment of social withdrawal in adolescents and adults with Fragile X Syndrome and for the treatment of autism spectrum disorders. AGI Therapeutics had been conducting clinical trials for the treatment of dyspepsia and for the treatment of delayed gastric emptying in diabetic patients; however, no recent development has been reported for this research. In 2015, Osmotica Pharmaceutical filed a NDA seeking approval of an extended-release formulation for the alleviation of spasticity due to multiple sclerosis.

AGI-006 is an oral formulation of arbaclofen, the R-isomer of baclofen. In 2012, a license option agreement was signed between Seaside and Roche by which the latter may commercialize the product upon completion of certain clinical development phases in fragile X syndrome and in autism spectrum disorders.

2D chemical structure of 1134-47-02D chemical structure of 1134-47-0Baclofen [USAN:USP:INN:BAN:JAN]
1134-47-0

2D chemical structure of 28311-31-1Baclofen hydrochloride
28311-31-1

2D chemical structure of 63701-55-3Arbaclofen hydrochloride
63701-55-3

2D chemical structure of 63701-56-4(S)-Baclofen hydrochloride
63701-56-4

2D chemical structure of 66514-99-6(S)-Baclofen
66514-99-6

2D chemical structure of 1395997-58-6Acamprosate mixture with baclofen
1395997-58-6

CLIP1

Strategy for asymmetric synthesis of (R)-(-)-Baclofen is as represented in the Scheme 14. Herein, we made use of asymmetric Michael addition of nitromethane to 4- Chlorochalcone in the presence of Cu(acac)2 and (-)-Sparteine as a catalyst in DCM for 8 h to provide γ-nitro ketone as colorless solid, mp 105-109°C, in 87% yield with 82% ee. The Michael adduct 3d on Baeyer-Villiger reaction using m-CPBA to produce corresponding nitro ester 6a. The reduction of 6a containing nitro group can be reduced with sodium borohydride in presence of NiCl2. It resulted to generate 7 cyclic pyrrolidine moiety in 65% yield. Which upon hydrolysis with HCl will lead to (R)-(-)- Baclofen 8 as a neurotransmitter inhibitor drug molecule

(R)-4-amino-3-(4-chlorophenyl)butanoic acid hydrochloride (8) The solution of 7 (100 mg, 0.51 mmol) in 6N HCl (2.7 mL) was refluxed at 100 °C. After 24 h, the reaction mixture was concentrated in vacuo to afford (R)-(–)- Baclofen 8 as colorless solid 93 mg, in 73% yield. Yield : 73% State : Solid. M.P. : 188-189 °C [a]D 25 : –3.4o (c = 0.65, H2O), lit.7 –3.79o (c = 0.65, H2O, 99 % ee) 1 H-NMR (300MHz, D2O) : δ. 7.36-7.49 (m, 4H) 3.50-3.37 (m, 2H), 2.30-3.22 (m, 1H), 2.71-2.92 (dd, 2H,) J = 9.5, 16.5 Hz).ppm 13C-NMR (75MHz, D2O) : δ. 175.46, 138.28, 136.95, 133.32, 129.32, 128.25, 127.81, 43.75, 39.91, 38.18.

7. Corey, E. J; Zhang, F. Y. Org. Lett. 2000, 2, 4257-4259

16. a) Thakur, V. V.; Nikalje, M. D.; Sudalai, A. Tetrahedron Asymmetry 2003, 14, 581. b) Chenevert R.; Desjardins, M.; Tetrahedron Lett. 1991, 32, 4249. c) Herdeis, C.; Hubmann, H. P. Tetrahedron Asymmetry 1992, 3, 1213. d) Meyers, A. I.; Snyder, L. J. Org. Chem. 1993, 58, 36.

clip 2

Yoshiji Takemoto (2005)6 Yoshiji Takemoto et al. have developed chiral thiourea catalyst 15 which was found to be highly efficient for the asymmetric Michael addition of 1,3-dicarbonyl compound to nitroolefins. Furthermore, a new synthetic route for (R)-(-)-Baclofen 14 and the generation of a chiral quaternary carbon center with high enantioselectivity by Michael reaction were developed (Scheme 6)

6. Okino, T.; Hoashi, Y.; Xuenong Xu,; Takemoto, Y.. J. Am. Chem. Soc. 2005, 127, 119.

CLIP3

Enantio- and Diastereoselective Michael Reaction of 1,3-Dicarbonyl Compounds to Nitroolefins Catalyzed by a Bifunctional Thiourea

Contribution from the Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
J. Am. Chem. Soc.2005127 (1), pp 119–125
DOI: 10.1021/ja044370p
Publication Date (Web): December 3, 2004
Copyright © 2005 American Chemical Society

Abstract

Abstract Image

We synthesized a new class of bifunctional catalysts bearing a thiourea moiety and an amino group on a chiral scaffold. Among them, thiourea 1e bearing 3,5-bis(trifluoromethyl)benzene and dimethylamino groups was revealed to be highly efficient for the asymmetric Michael reaction of 1,3-dicarbonyl compounds to nitroolefins. Furthermore, we have developed a new synthetic route for (R)-(−)-baclofen and a chiral quaternary carbon center with high enantioselectivity by Michael reaction. In these reactions, we assumed that a thiourea moiety and an amino group of the catalyst activates a nitroolefin and a 1,3-dicarbonyl compound, respectively, to afford the Michael adduct with high enantio- and diastereoselectivity.

http://pubs.acs.org/doi/full/10.1021/ja044370p

http://pubs.acs.org/doi/suppl/10.1021/ja044370p/suppl_file/ja044370psi20040916_090526.pdf

Synthesis of (R)()-Baclofen. γ-Amino butylic acid (GABA) plays an important role as an inhibitory neurotransmitter in the central nervous system (CNS) of mammalians,20,21 and the deficiency of GABA is associated with diseases that exhibit neuromuscular dysfunctions such as epilespy, Huntington’s and Parkinson’s diseases, etc.22 Baclofen is a lipophilic analogue of GABA, and it is widely used as an antispastic agent. Although baclofen is commercialized in its racemic form, it has been reported that its biological activity resides exlusively in the (R)-enantiomer.23 We next applied our enantioselective Michael reaction for the synthesis of (R)-(−)-baclofen (Scheme 1). The reaction of 4-chlorobenzaldehyde with nitromethane and subsequent dehydration of the resultant alcohol provided nitroolefin 9, which was reacted with diethyl malonate 3a in the presence of 10 mol % of 1e to afford the adduct 10 in 80% yield with 94% ee. Furthermore, enantiomerically pure 10 (>99% ee) was obtained after single recrystallization from Hexane/EtOAc. Reduction of the nitro group with nickel borite and in situ lactonization gave lactone 11 in 94%. The ester group of 11 was hydrolyzed and decarboxylated to afford 12. The specific rotation of 12 was compared with that of literature data24 ([α]30D −39.7° (c 1.00, EtOH), lit. [α]25D −39.0° (c 1, EtOH)), and, as expected, the absolute configuration of 12 was determined to be R. Lactam 12 was finally hydrolyzed with 6N HCl, affording enantiomerically pure (R)-(−)-baclofen as its hydrochloric salt with 38% overall yield in six steps from 4-chlorobenzaldehyde. Consequently, we succeeded in the synthesis of (R)-(−)-baclofen by the simple procedure with high enantioselctivity.

Figure

Scheme 1.  Total Synthesis of (R)-(−)-Baclofena

a Conditions:  (a) MeNO2, NaOMe, MeOH, room temperature, 15 h; (b) MsCl, TEA, THF, room temperature, 1 h; (c) diethyl malonate, 1e, toluene, room temperature, 24 h; (d) NiCl2·6H2O, NaBH4, MeOH, room temperature, 7.5 h; (e) NaOH, EtOH, room temperature, 45 h; (f) toluene, reflux, 6.5 h; (g) 6N HCl, reflux, 24 h.

Total synthesis of (R)-(–)-baclofen. 9: The mixture of 4-chlorobenzaldehyde (1.41 g, 10 mmol), nitromethane (10 equiv, 5.4 ml) and NaOMe (0.10 equiv, 54.0 mg) in MeOH (10 ml) was stirred overnight. Saturated ammonium chloride was added to the mixture and aqueous phase was extracted with AcOEt. The extract was washed with brine, dried over MgSO4, filtrated and concentrated in vacuo. The residue was purified by by column chromatography on silica gel (Hexane/AcOEt = 3/1 as eluent) to afford desired nitroalcohol 8 (1.82 g, 90%). To the stirred solution of the obtained nitroalcohol 8 and MsCl (1.2 equiv, 0.84 ml) in THF (9.0 ml) was added TEA (2.1 equiv, 2.7 ml) dropwise at 0 °C. After 1 h, saturated ammonium chloride was added to the reaction mixture and aqueous phase was extracted with AcOEt. The extract was washed with 1N HCl (two times), saturated NaHCO3 and brine, dried over MgSO4, filtrated and concentrated in vacuo. The residual solid was purified by recrystallization from AcOEt/Hexane to afford the desired nitroolefin 9 (1.20 g, 72%). yellow needle; m.p. 112 °C (AcOEt/Hexane); 1 H NMR (500 MHz, CDCl3) δ 7.97 (d, J = 13.7 Hz, 1H), 7.57 (d, J = 13.7 Hz, 1H), 7.50 (d, J = 8.6 Hz, 2H), 7.44 (d, J = 8.6 Hz, 2H) ppm; 13 C NMR (126 MHz, CDCl3) δ 138.4, 137.7, 137.5, 130.3, 129.8, 128.6 ppm; IR (CHCl3) ν 3113, 3029, 1637, 1594, 1525, 1494 cm-1 ; MS (EI + ) 183 (M+ , 51), 101 (100); Anal. Calcd. for C8H6ClNO2: C 52.34; H, 3.29; N, 7.63; Cl, 19.31. Found: C, 52.35; H, 3.40; N, 7.67; Cl, 19.24. 10: Under argon atmosphere, to the stirred solution of p-chloro-β-nitrostylene 9 (36.7 mg, 0.20 mmol) and thiourea (0.10 equiv, 8.3 mg) in toluene (0.40 ml) was added diethylmalonate (2 equiv, 0.060 ml) at rt. After 24 h, the reaction mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (AcOEt/hexane = 1/5 as eluent) to afford desired product 10 (55.3 mg, 80%) as colorless solid. Enantiomerically pure 10 (>99% ee) was obtained after single recrystallization from Hexane/AcOEt. m.p. 56-57 °C (Hexane/AcOEt); [α]D 25 –8.56 (c 1.02, CHCl3, >99% ee); 1 H NMR (500 MHz, CDCl3) δ 7.30 (d, J = 8.2 Hz, 2H), 7.19 (d, J = 8.6 Hz, 2H), 4.91 (dd, J = 4.6, 13.1 Hz, 1H), 4.83 (dd, J = 9.5, 13.1 Hz, 1H), 4.23 (m, 3H), 4.04 (q, J = 7.22 Hz, 2H), 3.78 (d, J = 9.5 Hz, 1H), 1.27 (t, J = 7.2 Hz, 3H), 1.09 (t, J = 7.0 Hz, 3H); 13 C NMR (126 MHz, CDCl3) δ 167.4, 166.8, 134.9, 134.5, 129.6, 129.3, 77.5, 62.3, 62.1, 54.8, 42.4, 14.0, 13.8 ppm; IR (CHCl3) ν 3031, 2994, 1733, 1558, 1494, 1374 cm-1 ; MS (FAB+ ) 344 (MH+ , 100); Anal. Calcd for C15H18ClNO6: C, 52.42, H, 5.28, N, 4.07, Cl, 10.31; Found: C, 52.52, H, 5.21, N, 4.07, Cl, 10.25; HPLC [Chiralcel OD-H, hexane/2-propannol = 90/10, 0.5 mL/min, λ = 210 nm, retention times: (major) 28.3 min, (minor) 25.1 min]. 11: Under argon atmosphere, to the suspension of 10 (550 mg, 1.60 mmol, >99% ee) and NiCl2· 6H2O (1.0 equiv, 380 mg) in MeOH (8.0 ml) was added NaBH4 (12 equiv, 726 mg) at 0 °C. After the reaction mixture was stirred 7.5 h at rt, the reaction mixture was quenched with NH4Cl and diluted with CHCl3. The organic layer was separated and dried over MgSO4, filtrated and concentrated in vacuo. The residue was purified by column chromatography on silica gel (MeOH/CHCl3 = 1/20 as eluent) to afford desired product (402 mg, 94%) as colorless powder. m.p. 126-128 °C (Hexane/AcOEt); [α]D 26 –123.4 (c 0.96, CHCl3); 1 H NMR (500 MHz, CDCl3) δ 7.31 (m, 2H), 7.20 (d, J = 8.2 Hz, 2H), 7.12 (s, 1H), 4.24 (q, J = 7.0 Hz, 1H), 4.09 (m, 1H), 3.81 (m, 2H), 3.54 (m, 1H), 3.41 (m, 1H), 1.28 (t, J = 6.9 Hz, 3H); 13 C NMR (126 MHz, CDCl3) δ 172.5, 169.0, 138.3, 133.5, 129.2, 128.4, 61.9, 55.2, 47.5, 43.7, 14.1 ppm; IR (CHCl3) ν 3435, 3229, 3017, 2360, 1710, 1493 cm-1 ; MS (FAB+ ) 268 (MH+ , 100); Anal. Calcd for C13H14ClNO3: C, 58.32, H, 5.27, N, 5.23, Cl, 13.24; Found: C, 58.10, H, 5.15, N, 5.43, Cl, 13.13. 12 : To the solution of 11 (240mg, 0.90 mmol) in EtOH (3.6 ml) was added 1N NaOH (1.1 ml) at rt. After 30 min, the reaction mixture was concerned in vacuo. To the residue was added H2O and 5N HCl, and the aqueous phase was extracted with CHCl3. The extract was dried over MgSO4, filtrated andconcentrated in vacuo to afford corresponding carboxylic acid (194 mg, 90%). The solution of carboxylic acid (194 mg, 0.81 mmol) in toluene (11 ml) was refluxed at 140 °C. After 6 h, the mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (MeOH/ CHCl3 = 1/7) to afford desired product 12 (148 mg, 93%) as colorless needle. m.p. 109-111 °C (Hexane/AcOEt); [α]D 30 –39.7 (c 1.00, CHCl3); 1 H NMR (500 MHz, CDCl3) δ 7.32 (d, J = 7.9 Hz, 2H), 7.19 (t, J = 8.2 Hz, 2H), 6.15 (s, 1H), 3.79 (t, J = 8.9 Hz, 1H), 3.68 (m, 1H), 3.38 (t, J = 8.4 Hz, 1H), 2.74 (dd, J = 9.0, 16.9 Hz, 1H), 2.45 (dd, J = 8.6, 16.8 Hz, 1H); 13 C NMR (126 MHz, CDCl3) δ 177.5, 140.7, 132.9, 129.0, 128.1, 49.3, 39.6, 37.8 ppm; IR (CHCl3) ν 3439, 3006, 2361, 1699, 1494 cm-1 ; MS (FAB+ ) 196 (MH+ , 100); Anal. Calcd for C10H10ClNO: C, 61.39, H, 5.15, N, 7.16, Cl, 18.12; Found: C, 61.50, H, 5.21, N, 7.25, Cl, 17.98. (R)-(–)-baclofen : The solution of 12 (107 mg, 0.55 mmol) in 6N HCl (2.7 ml) was refluxed at 100 °C. After 24 h, the reaction mixture was concentrated in vacuo to afford (R)-(–)-baclofen (129 mg, 94%) as colorless solid. m.p. 188-189 °C (exane/i-PrOH); [α]D 25 –3.79 (c 0.65, H2O); 1 H NMR (500 MHz, DMSO-d6) δ 12.26 (s, 1H), 8.13 (s, 3H), 7.35 (m, 4H), 3.09 (m, 1H), 2.94 (m, 1H), 2.85 (dd, J = 5.5, 16.2 Hz, 1H), 2.56 (dd, J = 9.5, 16.5 Hz, 1H); 13 C NMR (126 MHz, DMSO-d6) δ 172.5, 139.5, 131.9, 130.0, 128.7, 128.6, 128.0, 43.1, 39.1, 37.8 ppm; MS (FAB+ ) 214 (MH+ , 100); HRMS (FAB+ ) Calcd for [C10H13ClNO2] + : 214.0635; Found: 214.0637.

Image result for baclofen synthesis

http://www.sciencedirect.com/science/article/pii/S0957416604003672

Image result for baclofen synthesis

http://www.sciencedirect.com/science/article/pii/S0957416699002359

Image result for baclofen synthesisThe thiourea catalyst L7 bearing 3,5-bis(trifluoromethyl) benzene and dimethylamino groups has been revealed to be efficient for the asymmetric Michael reaction of 1,3-dicarbonyl compounds to nitroolefins (Scheme 8). This methodology has been applied for the total synthesis of (R)-(−)-baclofen. Reaction of 4-chloronitrostyrene and 1,3-dicarbonyl compound generates quaternary carbon center with 94% ee. Reduction of the nitro gruop to amine and subsequent cyclization, esterification and ring opening provides ( R )-(−)-baclofen in 38% yield.

Image result for baclofen synthesis

http://pubs.rsc.org/en/content/articlelanding/2010/np/b924964h/unauth#!divAbstract

Image result for baclofen synthesis

http://pubs.rsc.org/en/content/articlelanding/2010/np/b924964h/unauth#!divAbstract

Image result for baclofen synthesis

http://pubs.rsc.org/en/Content/ArticleHtml/2016/SC/c5sc02913a

Image result for baclofen synthesis

REF

Highly enantioselective biotransformations of 2-aryl-4-pentenenitriles, a novel chemoenzymatic approach to (R)-(-)-baclofen
Tetrahedron Lett 2002, 43(37): 6617

Enantioselective Michael addition of nitromethane to alpha,beta-enones catalyzed by chiral quaternary ammoniun salts. A simple synthesis of (R)-baclofen
Org Lett 2000, 2(26): 4257

Stereospecific synthesis of (R)- and (S)-baclofen and (R)- and (S)-PCPGABA [4-amino-2-(4chlorophenyl)butyric Acid] via (R)- and (S)-3-(4-Chlorophenyl)pyrrolidines
Chem Pharm Bull 1995, 43(8): 1302

Enantioselective syntheses of (-)-(R)-rolipram, (-)-(R)-baclofen and other GABA analogues via rhodium-catalyzed conjugate addition of arylboronic acids
Synthesis (Stuttgart) 2003, (18): 2805

Palladium-catalyzed, asymmetric Baeyer-Villiger oxidation of prochiral cyclobutanones with PHOX ligands
Tetrahedron 2011, 67(24): 4352

An efficient synthesis of (R)- and (S)-baclofen via desymmetrization
Tetrahedron Lett 2009, 50(45): 6166

Recoverable resin-supported pyridylamide ligand for microwave-accelerated molybdenum-catalyzed asymmetric allylic alkylations: Enantioselective synthesis of baclofen
Org Lett 2003, 5(13): 2275

Asymmetric synthesis of ß-substituted ?-lactams via rhodium/diene-catalyzed 1,4-additions: Application to the synthesis of (R)-baclofen and (R)-rolipram
Org Lett 2011, 13(4): 788

Multisite organic-inorganic hybrid catalysts for the direct sustainable synthesis of GABAergic drugs
Angew Chem Int Ed 2014, 53(33): 8687

///////////////

http://www.jocpr.com/articles/a-facile-synthesis-of-baclofean-via-feacac3-catalyzed-michael-addition-and-pinner-reaction.pdf

http://shodhganga.inflibnet.ac.in/bitstream/10603/93509/10/10_chapter1.pdf

Image result for baclofen synthesis

Image result for baclofen synthesis

Image result for baclofen synthesis

Image result for baclofen synthesis

Image result for baclofen synthesis

(±)-Baclofen, hydrochloride (2)

A mixture of 4-(4-Chlorophenyl) pyrrolidin-2-one 15 (0.070 g, 0.35 mmol) in HCl aqueous solution (6 mol L-1, 1.5 cm3) was heated at 100 °C for 6 h. The solvent was removed under reduced pressure and the residue was triturated in isopropanol yielding a crystalline (±)-baclofen hydrochloride 2 (0.071 g, 82%).; IR nmax/cm -1: 3415, 3006, 1713, 1562, 1492, 1407, 1251, 1186, 815 cm-1 (KBr, neat); 1H NMR (300 MHz, CDCl3d 2.55 (dd, J 16.5 and 8.7 Hz, 1 H); 2.82 (dd, J 16.5 and 5.7 Hz, 1 H); 2.93-3.50 (m, 3 H); 7.34 (d, J 8.7 Hz, 2 H), 7.40 (d, J 8.7 Hz, 2 H), 7.94 (bs, 3H, NH3+), 12.23 (bs, 1 H, COOH), 13C NMR (CDCl3, 75 MHz) d 37.94, 39.70, 43.28, 128.89, 130.27, 132.20, 139.56, 172.71.

http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0103-50532001000500011

Title: Baclofen
CAS Registry Number: 1134-47-0
CAS Name: b-(Aminomethyl)-4-chlorobenzenepropanoic acid
Additional Names: b-(aminomethyl)-p-chlorohydrocinnamic acid; g-amino-b-(p-chlorophenyl)butyric acid; b-(4-chlorophenyl)GABA
Manufacturers’ Codes: Ba-34647
Trademarks: Baclon (Leiras); Clofen (Alphapharm); Lioresal (Novartis)
Molecular Formula: C10H12ClNO2
Molecular Weight: 213.66
Percent Composition: C 56.21%, H 5.66%, Cl 16.59%, N 6.56%, O 14.98%
Literature References: Specific GABA-B receptor agonist. Prepn: NL 6407755; H. Keberle et al., US 3471548 (1965, 1969 both to Ciba). Toxicity study: T. Tadokoro et al., Osaka Daigaku Igaku Zasshi 28, 265 (1976), C.A. 88, 183016u (1978). Comprehensive description: S. Ahuja, Anal. Profiles Drug Subs. 14, 527-548 (1985). Review of pharmacology and therapeutic efficacy in spasticity: R. N. Brogden et al., Drugs 8, 1-14 (1974); of intrathecal use in spinal cord injury: K. S. Lewis, W. M. Mueller, Ann. Pharmacother.27, 767-774 (1993). Clinical evaluation in reflex sympathetic dystrophy: B. J. van Hilten et al., N. Engl. J. Med. 343, 625 (2000).
Properties: Crystals from water, mp 206-208° (Keberle); 189-191°, (Uchimaru). LD50 in male mice, rats (mg/kg): 45, 78 i.v.; 103, 115 s.c.; 200, 145 orally (Tadokoro).
Melting point: mp 206-208° (Keberle); 189-191°, (Uchimaru)
Toxicity data: LD50 in male mice, rats (mg/kg): 45, 78 i.v.; 103, 115 s.c.; 200, 145 orally (Tadokoro)
Derivative Type: Hydrochloride
Molecular Formula: C10H13Cl2NO2
Molecular Weight: 250.12
Percent Composition: C 48.02%, H 5.24%, Cl 28.35%, N 5.60%, O 12.79%
Properties: mp 179-181°.
Melting point: mp 179-181°
Therap-Cat: Muscle relaxant (skeletal).
Keywords: Muscle Relaxant (Skeletal).

/////////////////(R)-(–)-Baclofen, Arbaclofen, STX 209, AGI 006, Spasticity,  PREREGISTERD, OSMOTICA PHARMA

c1cc(ccc1[C@@H](CC(=O)O)CN)Cl


Filed under: Uncategorized Tagged: AGI-006, Arbaclofen, R-baclofen, STX-209

NNC 45-0781

$
0
0

Image result for NNC 45-0781

NNC 45-0781

Molecular Formula C27H29NO3
Molecular Weight 415.5241

CAS 207277-66-5

  • 2H-1-Benzopyran-7-ol, 3,4-dihydro-3-phenyl-4-[4-[2-(1-pyrrolidinyl)ethoxy]phenyl]-, cis-(-)-
  • (3S,4R)-3,4-Dihydro-3-phenyl-4-[4-[2-(1-pyrrolidinyl)ethoxy]phenyl]-2H-1-benzopyran-7-ol

2H-1-Benzopyran-7-ol, 3,4-dihydro-3-phenyl-4-(4-(2-(1-pyrrolidinyl)ethoxy)phenyl)-, (3S,4R)-

  • OriginatorNovo Nordisk
  • ClassOsteoporosis therapies; Pyrrolidines; Small molecules
  • Mechanism of ActionSelective estrogen receptor modulators

PATENT

WO 9818776

WO 9818771

WO 2003063859

A quantitative structure activity relationship study on cis-3,4-diaryl hydroxy chromones as high affinity partial agonists for the estrogen receptor
Chemistry: An Indian Journal (2003), 1, (3), 207-214

SYN 1

EP 0937057; WO 9818771, EP 0937060; WO 9818776

http://www.drugfuture.com/synth/syndata.aspx?ID=268276

Coumarin (III) was prepared by condensation of benzophenone (I) with phenylacetic acid (II) in the presence of Ac2O and Et3N. Reduction of the lactone function of (III) with LiAlH4, followed by acidic treatment furnished diaryl chromene (IV). Subsequent hydrogenation of (IV) over Pd/C gave rise to the racemic cis chromane (V), which was O-alkylated with 1-(2-chloroethyl) pyrrolidine (VI) producing the corresponding (pyrrolidinyl)ethoxy derivative. Resolution by means of active ditoluoyl tartaric acid yielded the desired (-)-enantiomer (VII). Finally, cleavage of the methoxy group using pyridine hydrochloride at 150 C provided the title compound.

PAPER

Bioorg Med Chem 2002,10(1),125

Abstract

The syntheses and in vitro pharmacological evaluation of a number of cis-3,4-diaryl-hydroxy-chromanes are reported, along with the results of a thorough in vivo profiling of the tissue-selective estrogen partial-agonist NNC 45-0781 [3, (−)-(3S,4R)-7-hydroxy-3-phenyl-4-(4-(2-pyrrolidinoethoxy)phenyl)chromane]. These studies showed that NNC 45-0781 is a very promising candidate for the prevention of post-menopausal osteoporosis, and the treatment of other health issues related to the loss of endogenous estrogen production.

The synthesis and pharmacological evaluation of a series of new tissue-selective estrogens, the cis-3,4-diaryl-hydroxy-chromanes, is described.

Unlabelled figure

 

 

(-)-(3S,4R)-7-Hydroxy-3-phenyl-4-(4-(2-pyrrolidinoethoxy)phenyl)chromane (3,=9a).

colorless powder 3, which contained 0.25 mol equiv of ethanol of crystallization; 0.90 g (27% yield),

mp 221–223 C.

1 H NMR (DMSOd6, 400 MHz) d: 1.60–1.73 (m, 4H), 2.40–2.50 (m, 4H), 2.69 (t, 2H), 3.47–3.57 (m, 1H), 3.92 (t, 2H), 4.14–4.25 (m, 2H), 4.32 (dd, 1H), 6.27 (dd, 1H), 6.30 (d, 1H), 6.44 (d, 2H), 6.60 (d, 2H), 6.65 (d, 1H), 6.70–6.80 (m, 2H), 7.09–7.20 (m, 3H), 9.25 (s, 1H).

MS (EI): 415 (M+), 84. HR-MS; calcd for C27H30NO3 (M+H+) 416.2225, found 416.2198. HR-MS; calcd for C28H32NO3 (M+H+) 430.2382, found 430.2376.

Chiral HPLC: Chiradex A, 5m, 2504 mm (Merck) column; eluent, 6:4 methanol/0.2% aqueous triethylammonium acetate buffer, pH=5.2; flow, 0.5 mL/min; UV 220 nm; Rt=19.2 min, >98%ee. Elemental analysis; calcd for C27H29NO3 0.25C2H5OH; C, 77.35; H, 7.20; N, 3.28%; found C, 77.39; H, 7.29; N, 3.12%. [a] 20 D=283 (c=1.004% in ethanol/3M HCl, 80:20). P.

 

PAPER

Abstract Image

A highly enantioselective method for quick access to dihydrocoumarins is reported. The reaction involves a cooperative catalytic process with carbene and in situ generated Brønsted acid as the catalysts. α-Chloro aldehyde and readily available and stable o-hydroxybenzhydryl amine substrates were used to generate reactive azolium ester enolate and ortho-quinone methide (o-QM) intermediates, respectively, to form dihydrocoumarins with exceptionally high diastereo- and enantioselectivities. The catalytic reaction products can be easily transformed to valuable pharmaceuticals and bioactive molecules.

Carbene and Acid Cooperative Catalytic Reactions of Aldehydes and o-Hydroxybenzhydryl Amines for Highly Enantioselective Access to Dihydrocoumarins

 Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
 Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, People’s Republic of China
Org. Lett., Article ASAP
DOI: 10.1021/acs.orglett.7b02883
Publication Date (Web): October 25, 2017
Copyright © 2017 American Chemical Society

/////////////NNC 45-0781

c1ccc(cc1)[C@H]2COc3cc(ccc3[C@H]2c4ccc(cc4)OCCN5CCCC5)O


Filed under: Uncategorized Tagged: NNC 45-0781

AD 35

$
0
0

str1

AD 35

IND-120499

MF C24 H27 N3 O3
Molecular Weight, 405.49
Spiro[cyclopropane-1,5′-[5H-1,3]dioxolo[4,5-f]isoindol]-7′(6′H)-one, 6′-[2-[1-(2-pyridinylmethyl)-4-piperidinyl]ethyl]-

6′-[2-[1-(2-Pyridinylmethyl)-4-piperidinyl]ethyl]spiro[cyclopropane-1,5′-[5H-1,3]dioxolo[4,5-f]isoindol]-7′(6’H)-one

1531586-58-9 CAS FREE FORM

1531586-64-7  PHOSPHATE

1531586-62-5  HYDROCHLORIDE

Zhejiang Hisun Pharmaceutical Co Ltd

Image result for Zhejiang Hisun Pharmaceutical Co Ltd

AD-35 is known to be a neuroprotectant, useful for treating Alzheimer’s diseases.

Zhejiang Hisun Pharmaceutical is developing an oral tablet formulation of AD-35, for treating Alzheimers disease . By August 2017, the phase I multiple doses trial had been completed in the US and would be completed in China soon

CAS 1531586-64-7  PHOSPHATE

6′-[2-[1-(Pyridin-2-ylmethyl)piperidin-4-yl]ethyl]spiro[cyclopropane-1,5′-[1,3]dioxolo[4,5-f]isoindol]-7′(6’H)-one phosphate

 Molecular Formula C24 H27 N3 O3 . H3 O4 P
 Molecular Weight 503.4847

With the rapid growth of the elderly population, the number of people suffering from Alzheimer’s disease (Alzheimer’s disease) also will be increased dramatically.Alzheimer’s disease is also known as Alzheimer-type dementia (Alzheimer type dementia), or the Alzheimer type senile dementia (senile dementia of the Alzheimer type). At present, although the prevalence of this disease on a global scale is still unknown, but according to the latest report from the US Alzheimer’s Association (the Alzheimer’s Association), and in 2011 the United States there are about 540 million people suffer from Alcatel the number of Alzheimer’s disease, and in 2050, in the United States suffering from the disease will increase to about 13.5 million. Therefore, the development of better efficacy and fewer side effects of new drugs to treat the disease it is a priority.

Alzheimer’s disease is the most common form of senile dementia, it has become the sixth leading cause of death of Americans, and 65 years and the fifth leading cause of death in Americans over 65 years. Although scientists have this disease carried out extensive and in-depth research, but so far, the exact cause of the disease remains unclear. Alzheimer’s disease is a progressive disease that continues to kill nerve cells, destroying nerve connections in the brain, resulting in brain tissue is damaged, leading to patients gradually lose memory, consciousness and judgment, and cause mood disorders and behavioral disorders in patients.

Alzheimer’s is an irreversible disease, and now there is no any drug can prevent the disease, and no drugs can cure the disease or slow the disease process. Drugs currently used to treat the disease can only alleviate or ameliorate symptoms of the disease. These drugs are FDA approved for use in the United States a total of five, four of which are acetylcholinesterase (acetylcholinesterase) inhibitors. Acetylcholine (acetylcholine) is a neurotransmitter, a chemical released by nerves, if produced in the brain acetylcholine system, i.e. damaged cholinergic system, it can result in associated with Alzheimer’s disease memory disorders; and acetylcholinesterase function is to catalyze the hydrolysis of acetylcholine, acetylcholine is decomposed. Because Alzheimer’s disease is accompanied

Attenuation of acetylcholine activity, thus inhibiting acetylcholinesterase is one way to treat this disease. As described above, in the present 5 treatment of Alzheimer’s disease drugs in clinical use, there are four acetylcholinesterase inhibitors, including acetylcholinesterase inhibitors such as donepezil (donepezil), tacrine (tacrine ), rivastigmine (rivastigmine), and galantamine (galantamine), wherein donepezil (Sugimoto et al US4895841 and 5100901;.. Pathi et al WO 2007077443;. Parthasaradhi et al WO 2005003092;. Dubey et al WO 2005076749; Gutman . et al WO 200009483;… Sugimoto et al J. Med Chem 1995, 38, 481) is a first-line treatment of Alzheimer’s disease drugs. However, donepezil and the other four drugs can only improve the patient’s symptoms, and this is the only improvement of symptoms is short, only lasting about 6-12 months, and the patient response rates to these drugs only about 50% (Alzheimer’s Association, 201 1 Alzheimer ‘Disease Facts and Figures, Alzheimer’s & Dementia, 201 1, 7 (2), 208). The present invention provides a new class of inhibitors of acetylcholinesterase, which is dioxole between a new class of derivatives of benzo, is more effective than donepezil and fewer side effects in the treatment of Alzheimer’s disease drug.

PATENT

WO 2014005421

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2014005421&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

Example 42: 6- [2- [l- (2-Pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxolo [4,5-f ] Isoindole-7, Γ-cyclopropane-5-one (Compound No. 1-29)

To the reaction flask was added 24.3 g (0.069 mol) of compound 11-5, 36.5 g (0.26 mol) of potassium carbonate, 243 ml of ethanol, 6.1 ml (0.044 mole) of triethylamine, heated to about 50 ° C, 0.049 mol) of 2-chloromethylpyridine hydrochloride was maintained at about 50 ° C for 5 hours. The reaction was complete and 750 ml of water was added. The solid was precipitated, filtered and the cake was washed with water and dried to give 17.8 g of compound 1-29. Rate: 63.4%. ‘HNMR (CDC13 . 3 ): [delta] 1.26 (dd, 2H, J = 6.1, 7.6 Hz), 1.35 (brs,. 3 H), 1.49-1.57 (m, 4H), 1.72 (D, 2H, J = 8.6Hz) (T, 2H, J = 7.9 Hz), 3.64 (s, 2H), 6.03 (s, 2H), 2.09 (t, 2H, J = 10.4 Hz), 2.89 (d, 2H, J = 10.7 Hz) , 7.42 (s, 1 H), 7.15 (dd, 1 H, J = 5.2, 6.7 Hz), 7.24 (s, 1 H), 7.41 (d, 1 H, J = 7.7 Hz), 7.64 (td, H, J = 7.6, 1.8 Hz), 8.55 (D,. 1 H, J = 4.2 Hz); the MS (ESI): m / Z 406 [m + H] + .

Example 46: 6- [2- [l- (2-Pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxolo [4,5-f ] Isoindole-7, Γ-cyclopropane] -5-one hydrochloride (Compound No. 1-33)

To the reaction flask was added 5 g (0.012 mol) of compound 1-29 and 25 ml of ethanol, heated at 50 ° C

(0.012 mol) of concentrated hydrochloric acid was added, and 1 g of activated charcoal was added to decolorize for 20 minutes. The filtrate was cooled to room temperature and 50 ml of isopropyl ether was added dropwise. The solid was precipitated, stirred for 1 hour, The ether cake was washed with ether and dried to give 5 g of compound 1-33 in a yield of 91.7%. Ethanol / isopropyl ether can be re-refined, the yield of about 90%. 1H-NMR is (D 2 0): 51.14 (T, 2 H, J-7.0 Hz), 1.38-1.70 (m,. 7 H), 1.96 (D, 2H, J = 13.3 Hz), 2.99-3.14 (m, H. 4 ), 3.50 (d, 2 H, J = 11.0 Hz), 4.37 (s, 2H), 5.93 (s, 2H), 6.28 (s, 1 H), 6.75 (s, 1 H), 7.47 (dd, J = 7.8, 1.7 Hz), 8.58 (d, 1 H, J = 4.4 Hz), 7.55 (d, 1 H, J = 7.8 Hz), 7.91 (td, ; MS (ESI): m / z 406 [M-Cl] & lt; + & gt ; .

Example 48: 6- [2- [l- (2-Pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxolo [4,5-f ] Isoindole-7, Γ-cyclopropan-5-one phosphate (Compound I-3S)

To the reaction flask was added 2 g (0.0049 mole) of compound 1-29 and 40 ml of ethanol, stirred at 60 ° C until all dissolved, 0.57 g (0.0049 mole) of 85% phosphoric acid was added, stirred and solidified,

Liter of ethyl acetate, cooled to room temperature, stirred for 1 hour, filtered, and a small amount of ethyl acetate was used to wash the filter cake and dried to give 2.1 g of compound 1-35 in a yield of 84.7%. 1H-NMR (D 2 0): δ 1.10 (t, 2 H, J = 7.2 Hz), 1.33-1.64 (m, 7 H), 1.92 (d, 2 H, J = 13.4 Hz), 2.95-3.09 (m, (S, 1 H), 6.69 (s, 1 H), 7.45 (s, 2 H), 4.34 (s, (d, 1 H, J-7.8 Hz), 7.88 (td, 1 H, J = 7.7, 1.2 Hz), 8.54 (d, 1 H, J = 4.6 Hz).

PATENT

CN 103524515

https://encrypted.google.com/patents/CN103524515B?cl=en

PATENT

CN 105859732

https://www.google.com/patents/CN105859732A?cl=en

Example 14: 6- [2- [l_ (2- pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxolo [4,5 -f] isoindole-7, prepared Γ- cyclopropane] phosphate 5-one (compound I) is

Figure CN105859732AD00182

[0146] Compound was added 2g (4.9 mmol) of formula XI to the reaction flask 50mL, 40mL of ethanol, 60 ~ 70 ° C dissolved by heating, added with stirring square. 57g 85% (4.9mmol) phosphoric acid, and the precipitated solid was added dropwise 40mL of acetic acid ethyl cooled to room temperature, stirred for 1 hour, filtered, the filter cake washed with a small amount of ethyl acetate, dried to give 2.3g white solid (compound I, HPLC purity: 99.8%). Yield: 92.7%, H bandit R (D2O): δ1 · l〇 (t, 2H, J = 7.2Hz), 1.33-1.64 (m, 7H), 1.92 (d, 2H, J = 13.4Hz), 2.95 -3.09 (m, 4H), 3.46 (d, 2H, J = 10.7Hz), 4.34 (s, 2H), 5.89 (s, 2H), 6.20 (s, 1H), 6.69 (s, 1H), 7.45 ( , 7.53 (d, lH, J 7.8Hz dd, lH, J = 5.2,7.4Hz) =), 7.88 (td, lH, J =

PATENT

WO 2017177816

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017177816&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText

Process for preparing AD-35 and its intermediates – comprising the reaction of a cyano ester with a Grignard reagent, followed by condensation and further manipulative steps.

A novel intermediate of AD-35 is claimed. Also claimed is a processes for preparing 6,7-dihydro-[1,3]dioxolo[4,5-f]isoindol-5-one comprising the reaction of a cyano ester compound in an isopropyl ester (Ti(i-Pr)4)) with a Grignard reagent in the presence of an ethyl magnesium halide. Further claimed are processes for preparing synthon of intermediates. A process for preparing a benzodioxole derivative, particularly AD-35 from intermediates is also claimed.

WO2014005421 reports a class of benzodioxole compounds, which have the activity to inhibit acetylcholinesterase and can be used to treat Alzheimer’s disease. Of these compounds, it is particularly noteworthy that 6- [2- [1- (2-pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxole And [4,5-f] isoindole-7,1′-cyclopropane] -5-one phosphate, codon AD-35, whose chemical structure is as follows:
AD-35 is a weaker acetylcholinesterase inhibitor that inhibits acetylcholinesterase activity in vitro is about one tenth of the activity of donepezil, but the compound exhibits comparable efficacy with donepezil in the Morris water maze test , That is, the effect of improving memory and learning ability is comparable to donepezil. This suggests that the AD-35 is likely to also have the effect of improving memory and learning through other mechanisms in the body. A further study of the rat model of Alzheimer’s disease induced by Aβ 25-35 found that AD-35 significantly inhibited the production and release of proinflammatory cytokines TNF-α and IL-1β, Small Aβ 25-35 on the nerve cell toxicity, effectively protect the nerve cells.
In addition, AD-35 also exhibits a certain ability to chelate transition metal ions such as Cu 2+ in vitro , while Cu 2+ accelerates the formation of Aβ fibers and enhances the toxicity of Aβ to neuronal cells, thereby promoting neuronal cell death , So excessive Cu 2+ in the brain is also considered to be one of the risk factors for Alzheimer’s disease (Sarell et al. J. Biol. Chem. 2010, 285 (53), 41533). From the chemical structure point of view, AD-35 molecules in the piperidine ring and pyridine ring on the two nitrogen atoms constitute a structural unit similar to ethylenediamine, which should be able to explain why this compound to a certain extent Chelating transition metal ions. In terms of the safety of the compounds, the acute toxicity of mice showed that the toxicity of AD-35 was much less than that of donepezil. A newly completed clinical single-dose incremental tolerance test (SAD) showed that the subjects taking 90 mg of AD-35 did not have any adverse effects at once, indicating that the compound was safe.
In summary, the AD-35 is promising to be a small side-effect drug for the treatment of Alzheimer’s disease, and its multiple mechanisms of action are likely to make this compound not only alleviate the symptoms of Alzheimer’s patients , And can delay the process of the disease.
Since the synthesis route of AD-35 and its analogs reported in WO2014005421 is too long, the operation is complicated and the yield is low, and some steps are not suitable for industrial production. Therefore, it is necessary to develop a new process route to overcome the above- Preparation method.
The preferred reaction conditions of the present invention are listed in the following schemes:
Step (1) :
Step (2) :
Step (3) :
Step (4) :
Step (5) :
Step (6) :
Step (7) :
Step (8) :

Specific implementation plan

The following examples are provided for the purpose of further illustrating the invention, but this is not intended to be limiting of the invention.
Reference Example 1: Preparation of the starting material of tert-butyl 4- [2- (p-toluenesulfonyloxy) ethyl] piperidine-1-carboxylate (Formula VIa)

[0103]

[0104]
To a 10 L reaction flask was added 800 g (3.49 mol) of tert-butyl 4- (2-hydroxyethyl) piperidine-1-carboxylate, 5 L of dichloromethane, 974 ml of (6.75 mol) of triethylamine and 16 g of 4-dimethyl (3L × 3), the organic phase was collected, dried over anhydrous sodium sulfate, and the reaction mixture was washed with anhydrous sodium sulfate , Filtered and the filtrate was concentrated under reduced pressure to give 1360.3 g of compound VIa (HPLC purity: 85%). 1 H NMR (DMSO-d 6 ): δ 0.85-0.93 (m, 2H), 1.38 (s, 9H), 1.42-1.52 (m, 5H), 2.43 (s, 3H), 2.59 (br s, 2H (D, 2H, J = 11.3 Hz), 4.05 (t, 2H, J = 6.1 Hz), 7.50 (d, 2H, J = 8.1 Hz), 7.79 (d, 2H, J = 8.3 Hz) MS (ESI): m / z 383 [M + Na] & lt; + & gt ; .
Reference Example 2: Preparation of the starting material 4- (2-iodoethyl) piperidine-1-carboxylate (Formula VIb)
To a 50 mL reaction flask was added 5 g (13.0 mmol) of tert-butyl 4- [2- (p-toluenesulfonyloxy) ethyl] piperidine-1-carboxylate (Formula VIa), 35 mL of acetone and 2.9 g (19.3 mmol The organic phase was washed with 50 mL of water. The organic phase was collected and the aqueous phase was extracted again with 50 mL of ethyl acetate. The organic phase was washed with 50 mL of water and extracted with 50 mL of water and 50 mL of water. The organic phases were combined, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to dryness to give 3.5 g of compound VIb in a yield of 79.5%. 1 H NMR (DMSO-d 6 ): δ 0.97-1.07 (m, 2H), 1.41 (s, 9H), 1.51-1.58 (m, 1H), 1.63-1.66 (m, 2H), 1.73-1.78 (m, 2H), 2.69 (br s, 2H), 3.31 (t, 2H, J = 7.3Hz), 3.96 (d, 2H, J = 10.3Hz); MS (ESI): m / + H] + .
Example 1: Preparation of 6-bromo-1,3-benzodioxole-5-carboxylic acid (Compound II)
To the 2L reaction flask, 100 g (0.60 mol) of piperine, 29 g (0.725 mol) of sodium hydroxide and 1 L of water were successively added, and 150 g (0.84 mol) of N-bromosuccinimide was added thereto, After the reaction was carried out for 45 min, the reaction was monitored by TLC. The reaction solution was concentrated dropwise with concentrated hydrochloric acid to adjust the pH of the reaction solution to 2 to 3, and the solid was precipitated. The ice was cooled, filtered and washed with water to obtain 117.4 g of compound II (HPLC purity: 82%), Yield 79.5%. 1 H NMR (DMSO-d 6 ): δ 6.15 (s, 2H), 7.30 (s, 1H), 7.32 (s, 1H), 13.17 (s, 1H).
Example 2: Preparation of 6-bromo-1,3-benzodioxole-5-carboxylic acid (Compound II)
To the 2L reaction flask, 100 g (0.60 mol) of piperine, 29 g (0.725 mol) of sodium hydroxide and 1 L of water were successively added, and 150 g (0.84 mol) of N-bromosuccinimide was added thereto, After the reaction was complete for 45 min, the reaction was monitored by TLC. After 1 L of ethyl acetate and 40 mL of concentrated hydrochloric acid were added, the mixture was stirred for 20 min. The organic phase was collected, concentrated to dryness, 200 mL of water and 600 mL of petroleum ether, stirred for 1 h, , And 116 g of compound II (HPLC purity: 92.0%) was dried to a yield of 78.9%. & Lt; 1 & gt ; H NMR data with Example 1.
Example 3: Preparation of ethyl 6-bromo-1,3-benzodioxole-5-carboxylate (Compound IIIa)
To a 2 L reaction flask was added 117.3 g (0.39 mol) of 6-bromo-1,3-benzodioxole-5-carboxylic acid (II), 585 mL of absolute ethanol, opened with a stirrer, (1.4mol) concentrated sulfuric acid, heating reflux reaction 6h, TLC monitoring reaction is completed. Water was added dropwise, and 1.2 L of water was added dropwise to remove the solid, filtered and washed with water, and dried at 35 to 45C to obtain 124.0 g of compound IIIa (HPLC purity: 85%) in a yield of 93.9%. . 1 H NMR (CDCl3 . 3 ): [delta] 1.39 (T, 3H, J = 7.1Hz), 4.34 (Q, 2H, J = 7.1Hz), 6.04 (S, 2H), 7.07 (S, IH), 7.31 ( s, 1H).
Example 4: Preparation of methyl 6-bromo-1,3-benzodioxole-5-carboxylate (Compound IIIb)
To a 1 L reaction flask was added 50 g (0.30 mol) of 6-bromo-1,3-benzodioxole-5-carboxylic acid (II), 500 mL of anhydrous methanol, opened with a stirrer, 33.3 mL (0.60 mol) of concentrated sulfuric acid was added dropwise and heated under reflux for 6 h. TLC test reaction is completed, ice water cooling, precipitation of solids, dropping 500mL of water, filtration, water washing filter cake, 45 ~ 55 ℃ drying 44.4 g compound IIIb, yield: 84.0%. 1 H NMR (DMSO-d 6 ): δ 3.83 (s, 3H), 6.19 (s, 2H), 7.35 (s, 1H), 7.36 (s, 1H).
Example 5: Preparation of 6-cyano-1,3-benzodioxole-5-carboxylate (Compound IVa)
To a 2 L reaction flask was charged 124 g (0.38 mol) of ethyl 6-bromo-1,3-benzodioxole-5-carboxylate (IIIa), 990 mL of N, N-dimethylformamide, After opening the stirrer, 33.1 g (0.09 mol) of potassium ferrocyanide and 103.3 g (0.54 mol) of cuprous iodide were added, heated to 120-140C for 5 h, and the TLC reaction was completed. Cooling, dropping water to precipitate a solid, filtering, and washing the filter cake. The filter cake was stirred in 1.9 L of dichloromethane for 30 min, filtered, the filtrate was added with 9 g of activated charcoal, decolorized for 30 min, filtered and the filtrate was concentrated to a small amount. The solid was precipitated, n-hexane was added dropwise, cooled with ice water, filtered and dried to give 82.8 g of compound IVa (HPLC purity: 99.5%), yield: 83.2%. . 1 H NMR (DMSO-D . 6 ): [delta] 1.34 (T, 3H, J = 7.1Hz), 4.33 (Q, 2H, J = 7.1Hz), 6.29 (S, 2H), 7.51 (S, IH), 7.57 (s, 1H).
Example 6: Preparation of 6-cyano-1,3-benzodioxole-5-carboxylate (Compound IVa)
To a 50 mL reaction flask was added 3.5 g (12.8 mmol) of ethyl 6-bromo-1,3-benzodioxole-5-carboxylate (IIIa), 35 mL of N, N-dimethylformamide , 2.3g (25.7mmol) cuprous cyanide, open stirring, 120 ~ 140 ℃ reaction 30 ~ 60min, TLC detection reaction is completed, cooling, dropping 30mL saturated ammonium chloride aqueous solution, precipitate solid, filter, water washing cake. The filter cake was dissolved in 200 mL of ethyl acetate and washed with saturated aqueous ammonium chloride (30 ml x 2 times). The organic phase was collected and the aqueous phase was extracted again with 100 ml of ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and filtered , And concentrated to give 2.0 g of compound IVa in a yield of 62.5%. & Lt; 1 & gt ; H NMR data with Example 5.
Example 7: Preparation of 6-cyano-1,3-benzodioxole-5-carboxylate (Compound IVb)
To a 1 L reaction flask was added 40 g (0.15 mol) of methyl 6-bromo-1,3-benzodioxole-5-carboxylate (IIIb), 11.4 g (31.0 mmol) of potassium ferrocyanide , 35.2 g (0.18 mol) of cuprous iodide, 240 mL of N, N-dimethylacetamide, 120 to 140 ° C in an oil bath for 2 to 3 hours, and the TLC reaction was completed. After cooling, 480 mL of water was added dropwise, Ice water cooling, filtration, water washing filter cake. Filter cake was dissolved in 500mL ethyl acetate and 200mL tetrahydrofuran mixture, heated to 80 ℃, adding 2g activated carbon, filtered, the filtrate was concentrated to a small amount, precipitation of solid, dropping 200mL petroleum ether, ice water cooling, filtration, petroleum ether washing filter The cake was dried to give 27.7 g of compound IVb in a yield of 87.6%. 1 H NMR (DMSO-d 6 ): δ 3.87 (s, 3H), 6.28 (s, 2H), 7.49 (s, 1H), 7.55 (s, 1H).
Example 8: Preparation of Spiro [6H- [1,3] dioxolo [4,5-f] isoindole-7,1′-cyclopropane] -5-one (Compound V)
To a 2 L reaction flask was added 16 g (0.072 mol) of compound of formula IVa, 160 mL of dichloromethane, stirred and dissolved under nitrogen. 24 mL (0.080 mol) of isopropyl tetrafis (4) isopropyl ether was added and cooled to 0 to 20 ° C A solution of 73 mL (0.22 mol) of ethylmagnesium bromide in diethyl ether (3M) was added and the reaction was complete after TLC. Slowly drop the water / tetrahydrofuran solution (64 mL water / 240 mL tetrahydrofuran), heat to 50 ° C, decalcinate with 2 g of activated charcoal and stir for 20 min. Filtration, ethyl acetate washing filter residue, the filtrate 40 ~ 50 ° C concentrated under reduced pressure, add 96mL ethyl acetate and 96mL water, stirring solid precipitation, dropping 290mL n-hexane, ice water cooling, filtration, n-hexane washing cake, Dried to give 11.9 g of compound V (HPLC purity: 70%) in a yield of 80.2%. 1 H NMR (DMSO-d 6 ): δ 1.33-1.41 (m, 4H), 6.11 (s, 2H), 6.86 (s, 1H), 7.09 (s, 1H), 8.53 (s, 1H).
Example 9: Preparation of Spiro [6H- [1,3] dioxolo [4,5-f] isoindole-7,1′-cyclopropane] -5-one (Compound V)
To a 500 mL reaction flask was added 10 g (48.8 mmol) of 6-cyano-1,3-benzodioxole-5-carboxylate (IVb), 200 mL of methyl tert-butyl ether, (50.7 mmol) of (IV) isopropyl ester was cooled to 0 to 20 ° C, and 49 mL (0.15 mol) of ethyl magnesium bromide in diethyl ether (3M) was slowly added dropwise. After completion of the drop, the TLC reaction was completed. (10 mL x 2 times), the organic phase was collected and the aqueous phase was extracted again with 100 mL of ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, and the activated charcoal was dried over 100 mL of ethyl acetate and extracted with 250 mL of ethyl acetate. Decolorization, filtration, the filtrate was concentrated to a small amount, dropping petroleum ether, ice water cooling, filtration, petroleum ether washing cake, drying 2.3g compound V, yield: 23.2%. & Lt; 1 & gt ; H NMR data with Example 8.
Example 10: 4- [2- (5-oxospiro [[1,3] dioxolo [4,5-f] isoindole-7,1′-cyclopropane] -6 Yl) ethyl] piperidine-1-carboxylate (Compound VIIa)
To a 250 mL reaction flask was added 11.9 g (0.041 mol) of compound of formula V, 84 mL of dimethylsulfoxide, 4 g (0.071 mol) of potassium hydroxide, 27.3 g (0.06 mol) of 4- [2- (p-toluenesulfonyloxy ) Ethyl] piperidine-1-carboxylate (Formula VIa), heated to 55-65 ° C for 3 to 4 hours, and the TLC reaction was completed. (150 mL x 2 times), the aqueous phase was extracted again with 200 mL of ethyl acetate, the organic phase was combined, and 3 g of activated charcoal was added to decolorize, stirred for 30 min, filtered, and the mixture was washed with 300 mL of ethyl acetate. The filtrate was concentrated to dryness under reduced pressure to give compound VIIa. 1 H NMR (CDCl 3 ): δ 1.08-1.19 (m, 2H), 1.28 (dd, 2H, J = 6.2, 7.4 Hz), 1.45 (s, 9H), 1.48-1.57 (m, 5H) (d, 2H, J = 12.7 Hz), 2.69 (t, 2H, J = 11.6 Hz), 3.20 (t, 2H, J = 7.6 Hz), 4.07 (d, 2H, J = 13.1 Hz) , 2H), 6.43 (S, IH), 7.23 (S, IH); the MS (ESI): m / Z 437 [m + of Na] + .
Example 11: 4- [2- (5-oxospiro [[1,3] dioxolo [4,5-f] isoindole-7,1′-cyclopropane] -6 Yl) ethyl] piperidine-1-carboxylate (Compound VIIa)
To a 250 mL reaction flask, 6.7 g (33.0 mmol) of compound of formula V, 100 mL of N, N-dimethylformamide, 2.6 g (65.0 mmol) of sodium hydroxide, 14 g (41.3 mmol) of 4- (2-iodoethyl ) Piperidine-1-carboxylic acid tert-butyl ester (VIb), 25-30 ° C for 1.5 h, TLC detection reaction was completed, 100 mL of water and 100 mL of ethyl acetate were added and the organic phase was washed with water (50 mL x 2 times) The organic phase was collected and the aqueous phase was extracted again with 100 mL of ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to dryness to give compound VIIa. & Lt; 1 & gt ; H NMR data with Example 10.
Example 12: 6- [2- (4-Piperidine) ethyl] spiro [[l, 3] dioxolo [4,5-f] isoindole- Propane] -5-one hydrochloride (Compound VIIIa)
To a 100 mL reaction flask was added the compound of formula VIIa obtained in Example 10, 30 mL of ethanol, 45 mL of ethyl acetate, 10.5 mL of concentrated hydrochloric acid. Open the stirrer, 50 ~ 60 ℃ reaction 3h, TLC detection reaction is completed, stop heating, ice water cooling, filtration, ethyl acetate detergent cake, drying, 8.5g off-white solid (compound VIIIa, HPLC purity: 97%) The Yield: 41.4% (calculated based on the amount of compound V in Example 10). 1 H NMR (D 2 O): δ 1.06 (t, 2H, J = 6.7Hz), 1.32-1.46 (m, 6H), 1.60 (m, 1H), 1.91 (d, 2H, J = 13.5Hz) (M, 4H), 3.39 (d, 2H, J = 12.8 Hz), 5.90 (s, 2H), 6.18 (s, 1H), 6.68 (s, 1H); MS (ESI): m / z 315 [M-Cl] + .
Example 13: 6- [2- [1- (2-Pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxolo [4,5-f ] Isoindole-7,1′-cyclopropane] -5-one (Compound XI)
A solution of 128.6 g (0.35 mol) of the compound of formula VIIIa, 90 g (0.54 mol) of 2-chloromethylpyridine hydrochloride (formula IXa), 965 mL of water, 26 g of activated carbon and 60 to 65C for 30 minutes were charged into a 2 L reaction flask, , And the residue was washed with 643 ml of water and 215 mL of ethanol. The solution was slowly added with 161 g (1.16 mol) of potassium carbonate. The reaction was carried out at 55 to 65 ° C for 4 to 5 hours. After completion of the TLC reaction, the reaction was cooled, filtered and dried to obtain 137 g of crude The crude product was dissolved in 1.37L ethanol and dissolved at 60-65 ° C. After decontamination with activated charcoal (27.4 g / times x 2 times), 4.11 L of water was added dropwise with stirring, the solid was precipitated, the ice was cooled, filtered, And dried to give 118.9 g of compound XI in 80% yield. 1 H NMR (CDCl 3 ): δ 1.26 (dd, 2H, J = 6.1, 7.6 Hz), 1.35 (br s, 3H), 1.49-1.57 (m, 4H), 1.72 (d, 2H, J = 8.6 (T, 2H, J = 7.9 Hz), 3.64 (s, 2H), 6.03 (s, & lt; RTI ID = 0.0 & gt; 2H), 6.42 (s, 1H), 7.15 (dd, 1H, J = 5.2, 6.7 Hz), 7.24 (s, 1H), 7.41 (d, 1H, J = 7.7 Hz), 7.64 (td, 7.6, 1.8 Hz =), 8.55 (D, IH, J = 4.2Hz); the MS (ESI): m / Z 406 [m + H] + .
Example 14: 6- [2- [1- (2-Pyridylmethyl) -4-piperidinyl] ethyl] spiro [[1,3] dioxolo [4,5-f ] Isoindole-7,1′-cyclopropane] -5-one phosphate (Compound I)
To a 50 mL reaction flask was added 2 g (4.9 mmol) of the compound of formula XI, 40 mL of ethanol, dissolved at 60-70 ° C and 0.57 g of 85% (4.9 mmol) of phosphoric acid was added with stirring. The solid was precipitated, 40 mL of ethyl acetate was added dropwise, To room temperature, stirred for 1 hour, filtered, a small amount of ethyl acetate to wash the filter cake, and dried to obtain 2.3 g of a white solid (Compound I, HPLC purity: 99.8%). Yield: 92.7%. 1 H NMR (D 2 O): δ 1.10 (t, 2H, J = 7.2Hz), 1.33-1.64 (m, 7H), 1.92 (d, 2H, J = 13.4Hz), 2.95-3.09 (m, 4H), 3.46 (d, 2H, J = 10.7 Hz), 4.34 (s, 2H), 5.89 (s, 2H), 6.20 (s, 1H), 6.69 (s, 1H), 7.45 (dd, 1H, J = 7.5, 7.4 Hz), 7.53 (d, 1H, J = 7.8 Hz), 7.88 (td, 1H, J = 7.7, 1.2 Hz), 8.54 (d, 1H, J = 4.6 Hz)
Multifunctional compound AD-35 improves cognitive impairment and attenuates the production of TNF-alpha and IL-1beta in an alphabeta25-35-induced rat model of alzheimer’s disease
J Alzheimer’s Dis 2017, 56(4): 1403
CN101626688A * Dec 11, 2007 Jan 13, 2010 雷维瓦药品公司 Compositions, synthesis, and methods of using indanone based cholinesterase inhibitors
WO2014005421A1 * Jul 3, 2013 Jan 9, 2014 Zhejiang Hisun Pharmaceutical Co., Ltd. Benzodioxole derivative and preparation method and use thereof
////////////Alzheimers disease, Zhejiang Hisun Pharmaceutical, AD 35, PHASE1, IND-120499
O=C5N(CCC2CCN(Cc1ccccn1)CC2)C3(CC3)c4cc6OCOc6cc45

Filed under: PHASE 1, PHASE1 Tagged: AD 35, Alzheimer's disease, IND-120499, PHASE1, Zhejiang Hisun Pharmaceutical

Novel lead compounds in pre-clinical development against African sleeping sickness

$
0
0

Med. Chem. Commun., 2017, 8,1872-1890
DOI: 10.1039/C7MD00280G, Review Article
Michael Berninger, Ines Schmidt, Alicia Ponte-Sucre, Ulrike Holzgrabe
This article reviews the recent progress in drug development against the African sleeping sickness.

Novel lead compounds in pre-clinical development against African sleeping sickness

 Author affiliations

Abstract

Human African trypanosomiasis (HAT), also known as African sleeping sickness, is caused by parasitic protozoa of the genus Trypanosoma. As the disease progresses, the parasites cross the blood brain barrier and are lethal for the patients if the disease is left untreated. Current therapies suffer from several drawbacks due to e.g. toxicity of the respective compounds or resistance to approved antitrypanosomal drugs. In this review, the different strategies of drug development against HAT are considered, namely the target-based approach, the phenotypic high throughput screening and the drug repurposing strategy. The most promising compounds emerging from these approaches entering an in vivo evaluation are mentioned herein. Of note, it may turn out to be difficult to confirm in vitro activity in an animal model of infection; however, possible reasons for the missing efficacy in unsuccessful in vivo studies are discussed.

Conclusion  There are various starting points to generate hit compounds for the treatment of  African sleeping sickness. Especially stage II of HAT which is very hard to treat poses a  tough challenge for drug discovery programs as molecules inevitably need to cross the BBB. However, promising compounds (2, 15, and 17) are in the pipeline accomplishing these criteria for CNS mouse models, and in some cases even are  orally bioavailable (15 and 17). Especially the large phenotypic screening campaigns performed by the GNF, GlaxoSmithKline, DDU, and Sykes et al. resulted in promising hits discussed herein. Nevertheless, it is not always easy to translate results from in vitro studies into in vivo efficacy like shown in several of the mentioned studies. The reasons for in vivo failures are multilayered and might originate from (I) extensive  metabolism, (II) high plasma protein binding, (III) poor water solubility, (IV) efflux  transporters, (V) different sensitivity for particular strains, (VI) reduced permeability,  and (VII) growth inhibition rather than trypanocidal effects.

Image result for University of Würzburg Ulrike Holzgrabe

  • 1974 – 1981
    Studied chemistry and pharmacy at Marburg University and Kiel University
  • 1990 – 1999
    C3 professor at the University of Bonn, Germany
  • 1994 – 1995
    Visiting professor at the University of Erlangen-Nuremberg, Germany, and the University of Illinois at Chicago, USA
  • 1997 – 1999
    Vice-rector for teaching, studies and study reform at the University of Bonn
  • Since 1999
    C4/W3 professor of pharmaceutical chemistry at the University of Würzburg, Germany
  • Since 2009
    Dean of the Faculty of Chemistry and Pharmacy at the University of Würzburg

 Selected publications

  • Mohr, K. et al.: Rational design of dualsteric GPCR ligands: quests and promise. In: Br. J. Pharmacol. 159, 2010. pp. 997-1008.
  • Antony, J. et al.: Dualsteric GPCR targeting: a novel route to binding and signalling pathway selectivity. In: FASEB J. 23, 2009. pp. 442-450 (Listed as a “Must Read” by the “Faculty of 1000 Biology – the expert guide to the most important advances in biology”).
  • Niedermeier, S. et al.: A small-molecule inhibitor of Nipah virus envelope protein-mediated membrane fusion. In: J. Med. Chem. 52, 2009. pp. 4257-4265.
  • Göbel, T. et al.: In search of novel agents for therapy of tropical diseases and human immunodeficiency virus. In: J. Med. Chem. 51, 2008. pp. 238-250.
  • Hörr, V. et al.: Laser-induced fluorescence-capillary electrophoresis and fluorescence microplate reader measurement: two methods to quantify the effect of antibiotics. In: Anal. Chem. 79, 2007. pp. 7510-7518 (reviewed by D.L. Shenkenberg in Biophotonics International, Dec. 2007, pp. 57-58).
  • Disingrini, T. et al.: Design, synthesis, and action of oxotremorine-related hybrid-type allosteric modulators of muscarinic acetylcholine receptors. In: J. Med. Chem. 49, 2006. pp. 366-372.

 Selected projects

  • Characterisation of the oncogenic signalling network in multiple myeloma: development of targeted therapies, clinical research group KFO 216, inhibitors of the HSF/HSP system for treating multiple myeloma, since 2009
  • Identification, preparation and functional analysis of active ingredients for combating infectious diseases, SFB 630, small molecules for treating tropical infectious diseases, since 2003
  • Allosteric modulators and subtype-selective ligands of the muscarinic receptors, since 1991

 Membership in scientific bodies/juries

  • German Research Foundation (DFG) review-board member at the University of Würzburg, Germany, since 2009
  • Member of the Board of Pharmaceutical Science, International Federation of Pharmacy (FIP), since 2008
  • Member of the executive committee, European Federation for Pharmaceutical Sciences (Eufeps), since 2007
  • President of the German Pharmaceutical Society, 2004 – 2007
  • Member of the board of trustees of the University of Bonn, Germany, 2003 – 2007
  • Member of the scientific advisory board, German Federal Institute for Drugs and Medical Devices (BfArM), since 2002
  • Member of the German and European pharmacopoeia commissions, as well as president of several German and European pharmacopoeia boards, since 2001
 Image result for University of Würzburg Michael Berninger
Image result for University of Würzburg Michael Berninger
Image result for University of Würzburg Michael Berninger
Image result for University of Würzburg Institute of Pharmacy and Food Chemistry
WURZBERG
Image result for University of Würzburg Institute of Pharmacy and Food Chemistry
Image result for University of Würzburg Institute of Pharmacy and Food Chemistry
Image result for University of Würzburg Institute of Pharmacy and Food Chemistry
///////////University of Würzburg,  Ulrike Holzgrabe

Filed under: Uncategorized Tagged: African Sleeping Sickness, Ulrike Holzgrabe, University of Würzburg

BMS-986020

$
0
0

imgImage result for BMS-986020

BMS-986020

AM-152; BMS-986020; BMS-986202

cas 1257213-50-5
Chemical Formula: C29H26N2O5
Molecular Weight: 482.536

(R)-1-(4′-(3-methyl-4-(((1-phenylethoxy)carbonyl)amino)isoxazol-5-yl)-[1,1′-biphenyl]-4-yl)cyclopropane-1-carboxylic acid

Cyclopropanecarboxylic acid, 1-(4′-(3-methyl-4-((((1R)-1-phenylethoxy)carbonyl)amino)-5-isoxazolyl)(1,1′-biphenyl)-4-yl)-

1-(4′-(3-Methyl-4-(((((R)-1-phenylethyl)oxy)carbonyl)amino)isoxazol-5-yl)biphenyl-4-yl)cyclopropanecarboxylic acid

UNII: 38CTP01B4L

For treatment for pulmonary fibrosis, phase 2, The lysophosphatidic acid receptor, LPA1, has been implicated as a therapeutic target for fibrotic disorders

Lysophospholipids (LPs), including lysophosphatidic acid (LPA), sphingosine 1-phospate (S1P), lysophosphatidylinositol (LPI), and lysophosphatidylserine (LysoPS), are bioactive lipids that transduce signals through their specific cell-surface G protein-coupled receptors, LPA1-6, S1P1-5, LPI1, and LysoPS1-3, respectively. These LPs and their receptors have been implicated in both physiological and pathophysiological processes such as autoimmune diseases, neurodegenerative diseases, fibrosis, pain, cancer, inflammation, metabolic syndrome, bone formation, fertility, organismal development, and other effects on most organ systems.

Image result for Amira Pharmaceuticals

  • Originator Amira Pharmaceuticals
  • DeveloperB ristol-Myers Squibb; Duke University
  • Class Antifibrotics; Azabicyclo compounds; Carboxylic acids; Small molecules; Tetrazoles
  • Mechanism of Action Lysophosphatidic acid receptor antagonists
  • Orphan Drug Status Yes – Fibrosis
  • Phase II Idiopathic pulmonary fibrosis
  • Phase IPsoriasis

Most Recent Events

  • 05 May 2016 Bristol-Myers Squibb plans a phase I trial for Psoriasis in Australia (PO, Capsule, Liquid) (NCT02763969)
  • 01 May 2016 Preclinical trials in Psoriasis in USA (PO) before May 2016
  • 14 Mar 2016 Bristol-Myers Squibb withdraws a phase II trial for Systemic scleroderma in USA, Canada, Poland and United Kingdom (PO) (NCT02588625)

BMS-986020, also known as AM152 and AP-3152 free acid, is a potent and selective LPA1 antagonist. BMS-986020 is in Phase 2 clinical development for treating idiopathic pulmonary fibrosis. BMS-986020 selectively inhibits the LPA receptor, which is involved in binding of the signaling molecule lysophosphatidic acid, which in turn is involved in a host of diverse biological functions like cell proliferation, platelet aggregation, smooth muscle contraction, chemotaxis, and tumor cell invasion, among others

Image result for BMS-986020

PRODUCT PATENT

GB 2470833, US 20100311799, WO 2010141761

Hutchinson, John Howard; Seiders, Thomas Jon; Wang, Bowei; Arruda, Jeannie M.; Roppe, Jeffrey Roger; Parr, Timothy

Assignee: Amira Pharmaceuticals Inc, USA

Image result for Hutchinson, John Howard AMIRA

John Hutchinson

PATENTS

WO 2011159632

WO 2011159635

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013025733&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

WO 2013025733

Synthesis of Compound 74

Synthetic Route (Scheme XLV)

Compound 74 Compound 74a

[0562] Compound XLV-1 was prepared by the same method as described in the synthesis of compound 1-4 (Scheme 1-A).

[0563] To a solution of compound XLV-1 (8 g, 28.08 mmol) in dry toluene (150 mL) was added compound XLV-2 (1.58 g, 10.1 mmol), triethylamine (8.0 mL) and DPPA (9.2 g, 33.6 mmol). The reaction mixture was heated to 80 °C for 3 hours. The mixture was diluted with EtOAc (50 mL), washed with brine, dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (PE/EA = 10 IX) to give compound XLV-3 (9.4 g, yield: 83 %). MS (ESI) m/z (M+H)+402.0.

[0564] Compound 74 was prepared analogously to the procedure described in the synthesis of Compound 28 and was carried through without further characterization.

[0565] Compound 74a was prepared analogously to the procedure described in the synthesis of Compound 44a. Compound 74a: 1HNMR (DMSO-d6 400MHz) δ 7.81 (d, J = 8.4 Hz, 2H), 7.41 (d, J = 8.4 Hz, 2H), 7.52 (d, J = 8.4 Hz, 2H), 7.29-7.32 (m, 7 H), 5.78 (q, 1 H), 2.15 (s, 3 H), 1.52 (d, J = 6.0 Hz, 3H), 1.28 (br, 2 H), 0.74 (br, 2 H). MS (ESI) m/z (M+H)+ 483.1.

Paper

Development of a Concise Multikilogram Synthesis of LPA-1 Antagonist BMS-986020 via a Tandem Borylation–Suzuki Procedure

Chemical and Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00301

http://pubs.acs.org/doi/10.1021/acs.oprd.7b00301

Abstract Image

The process development for the synthesis of BMS-986020 (1) via a palladium catalyzed tandem borylation/Suzuki reaction is described. Evaluation of conditions culminated in an efficient borylation procedure using tetrahydroxydiboron followed by a tandem Suzuki reaction employing the same commercially available palladium catalyst for both steps. This methodology addressed shortcomings of early synthetic routes and was ultimately used for the multikilogram scale synthesis of the active pharmaceutical ingredient 1. Further evaluation of the borylation reaction showed useful reactivity with a range of substituted aryl bromides and iodides as coupling partners. These findings represent a practical, efficient, mild, and scalable method for borylation.

1H NMR (500 MHz, DMSO-d6) δ 1.19 (dd, J = 6.8, 3.8 Hz, 2H), 1.50 (dd, J = 6.8, 3.8 Hz, 2H), 1.56 (br s, 3H), 2.14 (br s, 3H), 5.78 (br s, 1H), 6.9–7.45 (br, 5H), 7.45 (br d, J = 8.3 Hz, 2H), 7.65 (d, J = 8.3 Hz, 2H), 7.79 (br d, 2H), 7.82 (br d, 2H), 8.87 (br s, 0.8H), 9.29 (s, 0.2H), 12.39 (br s, 1H). 13C NMR (126 MHz, DMSO-d6) δ 9.2, 15.8, 22.4, 28.3, 72.8, 113.8, 125.4, 125.6, 126.2, 126.3, 127.1, 127.7, 128.4, 130.9, 137.4, 140.0, 141.5, 142.2, 154.4, 159.6, 160.8, 175.2. HRMS (ESI+) Calculated M + H 483.19145, found 483.19095.

REFERENCES

1: Kihara Y, Mizuno H, Chun J. Lysophospholipid receptors in drug discovery. Exp
Cell Res. 2015 May 1;333(2):171-7. doi: 10.1016/j.yexcr.2014.11.020. Epub 2014
Dec 8. Review. PubMed PMID: 25499971; PubMed Central PMCID: PMC4408218.

//////////////BMS-986020,  AM 152, BMS 986020, BMS 986202, Orphan Drug, BMS, Amira Pharmaceuticals, Bristol-Myers Squibb, Duke University, Antifibrotics, PHASE 2, pulmonary fibrosis

O=C(C1(C2=CC=C(C3=CC=C(C4=C(NC(O[C@H](C)C5=CC=CC=C5)=O)C(C)=NO4)C=C3)C=C2)CC1)O


Filed under: 0rphan drug status, Phase2 drugs Tagged: AM 152, Amira Pharmaceuticals, Antifibrotics, bms, BMS 986202, BMS-986020, Bristol-Myers Squibb, Duke University, Orphan Drug, phase 2, pulmonary fibrosis

Enclomiphene citrate, New patent, WO 2017182097, F.I.S. – FABBRICA ITALIANA SINTETICI S.P.A

$
0
0

Image result for F.I.S. - FABBRICA ITALIANA SINTETICI S.P.A

Enclomiphene citrate, New patent, WO 2017182097, F.I.S. – FABBRICA ITALIANA SINTETICI S.P.A

WO-2017182097

F.I.S. – FABBRICA ITALIANA SINTETICI S.P.A

CARUANA, Lorenzo; (IT).
PADOVAN, Pierluigi; (IT).
DAL SANTO, Claudio; (IT)

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017182097&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

Image result for F.I.S. - FABBRICA ITALIANA SINTETICI S.P.A

Enclomiphene citrate is an active pharmaceutical ingredient currently under evaluation in clinical phase III for the treatment of secondary hypergonadism. Moreover, it also could be potentially used for an adjuvant therapy in hypogonadal men with Type 2 diabetes.

Enclomiphene citrate of formula (I):

has chemical name of Ethanamine, 2-[4-[(1 )-2-chloro-1 ,2-diphenyl ethenyl]phenoxy]-/V,/V-diethyl-, 2-hydroxy-1 ,2,3-propanetricarboxylate (1 : 1 ); has CAS RN. 7599-79-3, and it is also named trans-Clomiphene monocitrate, E-Clomiphene citrate or Enclomiphene monocitrate.

Enclomiphene is component of Clomiphene, an active pharmaceutical ingredient, having chemical name Ethanamine, 2-[4-(2-chloro-1 ,2- diphenylethenyl)phenoxy]-N,N-diethyl, since Clomiphene is a mixture of the geometric isomers trans-Clomiphene (i.e. Enclomiphene) and cis- Clomiphene.

The US patent 3,848,030, in examples 31 and 32, discloses a process for the resolution of the geometric isomers of Clomiphene through the preparation of salts with racemic binaphthyl-phosphoric acid.

In the later publication Acta Cryst. (1976), B32, pag. 291 -293, the actual geometric isomery has been definitely established by single crystal X-Ray diffraction.

Finally, in the publication “Analytical profiles of drug substances and excipients”, vol. 25, (1998), pag. 85-121 , in particular at pag. 99, it is stated that prior to 1976 the cis stereochemistry was wrongly assigned to the trans-isomer of Clomiphene (E-Chlomiphene or Enclomiphene), and only after the above publication on Acta Cryst. the correct geometric isomery has been definitively assigned.

These observations in the prior art have been confirmed by our experimentation. In particular, repeating the experiment 31 of US patent 3,848,030, the trans-Clomiphene salt with racemic binaphthyl-phosphoric acid was isolated and not the salt with cis-Clomiphene as stated in said patent, as confirmed by 2D H-NMR analysis (NOESY experiment). Thus, Example 31 of US3,848,030, provides, at the end, Enclomiphene citrate, crystallized from a mixture of ethyl ether and ethanol, having a m.p. of 133-135°C. Example 32, instead provided Cis-Clomiphene citrate, crystallized from a mixture of ethyl ether and ethanol, having a m.p. of 120-126°C.

Thus, with the aim of preparing Enclomiphene citrate, whole experiment 31 of US3,848,030 has been reworked also carrying out the crystallization of the product form a mixture of ethyl ether and ethanol, hence providing a not crystalline solid with two DSC peaks respectively at 1 14°C and 188°C, although the starting material used for the reworking example was quite a pure substance (HPLC Analysis (A A%) is 98.95% of Enclomiphene), and having a substantially the same chemical purity of that used in the prior art experiment (m.p. of our Enclomiphene BPA salt was 218°C versus 220- 222°C of the prior art Enclomiphene BPA salt of Example 31 ).

The patent US2,914,563, in example 3, discloses a process for the preparation of trans-Clomiphene citrate, containing from 30% to 50% of cis-Clomiphene, as citrate, by reaction of 1 -ρ-(β- diethylaminoethoxy)phenyl]-1 ,2-diphenylethylene hydrochloride with N- chlorosuccinimmide in dry chloroform under reflux.

Khimiko-Farmatsevticheskii Zhurnal (1984), 18(1 1 ), 1318-24 English translation in the review Pharmaceutical Chemistry Journal November 1984, Volume 18, Issue 1 1 , pag. 758-764 (Title: Synthesis and biological study of the cis- and trans-isomers of Clomiphene citrate and some intermediates of its synthesis) discloses the trans-isomer of Clomiphene citrate, i.e. Enclomiphene citrate, characterized by:

1 H-NMR (MeOD) d 7.4-6.7 (m, 14H); 4.27 (t, 2H, -OCH2); 3.51 (t, 2H, CH2- N); 3.28 (q, 4H, 2xN-CH2)); 2.73 (2H); 2.78 (2H); 1.31 (t, 6H, 2xN-C-CHs)) Melting point: 138-139°C (98% purity by GLC);

IR spectrum, v cm-1 (suspension in mineral oil): 3640, 3430, 1720, 1710

(citrate), 1600-1555 (broad band, stilbene system); 750.

UV spectrum: λ max = 243 nm, ε 21 ,800 and λ max 300 nm, ε 1 1 ,400.

These prior art methods for the preparation of Enclomiphene citrate do not allow the preparation of Enclomiphene citrate having needle shaped crystal habit, indeed the crystallization by means of a mixture of ethyl ether and ethanol does not provide a crystalline solid having needle crystals.

Moreover, Enclomiphene citrate was described in literature with different melting points, in particular, 133-135°C and 138-139°C. Said solid forms of Enclomiphene citrate fail to comply with stabilities studies and furthermore show relatively poor solubility in water either in neutral or acid pH.

Furthermore, the prior art methods have the drawbacks related to the poor reproducibility of the process and of the solid form thus obtained.

Image result for F.I.S. - FABBRICA ITALIANA SINTETICI S.P.A

Image result for F.I.S. - FABBRICA ITALIANA SINTETICI S.P.A

EXPERIMENTAL SECTION

The starting material Clomiphene citrate can be prepared according to well-known prior art methods, or for example, as described in the example 1 of PCT/EP2015/074746 or can be purchased on the market.

[00190] Example 1 : Preparation of salt of Enclomiphene with racemic binaphthyl- phosphoric acid, starting from Clomiphene citrate.

Clomiphene citrate

[00191] A round bottom flask was charged 100 gr of Clomiphene Citrate (HPLC analysis (A/A%): 65.21 % Enclomiphene, 34.06% Z-Clomiphene) and 1000 mL of methanol. The suspension was stirred at 30°C up the complete

dissolution. Then a solution of racemic binaphthyl-phosphoric acid (abbreviated BPA) 30 gr (0.515 eq) in 30 ml_ of DMF was added. At the end of addition the mixture was stirred for 1 h at 30°C. The obtained suspension was filtered and the solid was washed with 100 ml_ of methanol.

[00192] 50.4 gr of Enclomiphene BPA salt (III) were obtained.

[00193] HPLC Analysis (A/A%): 97.04% Enchlomiphene, 2.5% Z-Clomiphene.

[00194] Example 1 b: Preparation of salt of Enclomiphene with racemic binaphthyl- phosphoric acid, starting from Clomiphene citrate.

[00195] A round bottom flask was charged 50 gr of Clomiphene Citrate and 500 ml_ of methanol. The suspension was heated at 40-45°C and stirred up to the complete dissolution. Then a solution of BPA 15 gr (0.515 eq) in 300 ml_ of methanol was added. At the end of addition the mixture was stirred for 1 h at 20°C. The obtained suspension was filtered and the solid was washed with 100 ml_ of methanol.

24.1 gr of Enclomiphene BPA salt were obtained.

HPLC Analysis (A/A%): 98.96% Enchlomiphene, 0.69% Z-Clomiphene.

[00196] Example 1 c: Preparation of salt of Enclomiphene with racemic binaphthyl- phosphoric acid, starting from Clomiphene citrate.

[00197] In a round bottom flask was charged 100 gr of Clomiphene Citrate and 1000 ml_ of methanol. The suspension was heated at 40-45°C and stirred up the complete dissolution. Then a solution of BPA 30 gr (0.515 eq) in 1000 ml_ of methanol was added. At the end of addition the mixture was stirred for 1 h at 20°C. the obtained suspension was filtered and the solid was wash with 100 ml_ of methanol.

47.9 gr of Enclomiphene BPA salt were obtained.

HPLC Analysis (A/A%): 98.81 % Enclomiphene, 0.79% Z-Clomiphene.

[00198] Example 1d: Preparation of salt of Enclomiphene with racemic binaphthyl- phosphoric, starting from Clomiphene citrate.

[00199] In a round bottom flask was charged 150 gr of Clomiphene citrate and 1500 mL of methanol. The suspension was heater at 40-45°C and stirred up the complete dissolution. Then a solution of BPA 45 gr (0.515 eq) in 900 mL of methanol was added. At the end of addition the mixture was

stirred for 1 h at 20°C. the obtained suspension was filtered and the solid was wash with 100 ml_ of methanol.

76.4 gr of E-Clomiphene BPA salt were obtained.

HPLC Analysis (A/A%): 98.82% Enchlomiphene, 0.80% Z-Clomiphene.

[00200] Example 2: Recrystallization of Enclomiphene BPA salt of formula (III) (the step A).

(Ill)

[00201] Into a proper 0.5 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene BPA salt (III) (50 g) and having Z-isomer of 1.64 % was suspended in DMF (2.1 L/Kg of Enclomiphene BPA (III)) and methanol (1.4 L/Kg of Enclomiphene BPA salt (III)). The suspension was heated to reflux (~ 76-79°C). Further DMF (0.1 L/Kg of Enclomiphene BPA (III)) might be required to improve the solubility of the starting material. Once the starting material was completely dissolved, methanol was added as anti-solvent (3.5 L/Kg of Enclomiphene BPA (III)). The temperature was decreased to 60°C and the mixture was stirred for 2 – 3 h. Then, the temperature was further decreased to 20 °C and filtered. The wet cake was washed twice with methanol (1.5 L/Kg of Enclomiphene BPA salt (III)). The product was dried under vacuum at 60 – 70 °C for 12 – 24 h. Time of drying could be prolonged until residual DMF is < 2500 ppm.

[00202] Analysis of quality of the final product of the above mentioned example and of the same product, obtained from repetition following the same process, it is shown in the following table:

Enclomiphene BPA (III) salt Enclomiphene BPA (III) salt rixx (Starting product) (finale product)

Z-isomer = 1.64 A/A% Z-isomer = 0.07 A/A%

Z-isomer = 0.79 A/A% Z- isomer = 0.03 A/A%

[00203] Example 3: Preparation of Enclomiphene citrate of formula (I), having needle shaped crystal habit, starting from Enclomiphene BPA salt formula (III).

(II)

[00204] Into a proper 4 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene BPA salt of formula (III) (400 g, assay 99.8 wt% 0.528 mol, 1 equiv.) was suspended in methyl-tert-butyl ether (MTBE, 2 L), isopropanol (IPA, 0.5 L) and water (2 L). The mixture was stirred for 15 minutes, then 0.48 L of ammonia solution 30 wt% was added and the mixture was further stirred for one hour. The aqueous phase was separated and the organic layer was washed with a solution of ammonia solution 30 wt% (0.12 L) and water (0.6 L). The aqueous phase was separated and the organic layer was finally washed with water (0.6 L). The organic solution was evaporated to residue under vacuum at 60-65°C. The residue was dissolved in 1.36 L of absolute ethanol. The assay of the solution was determined at this stage through a potentiometric titration and results in 15.125 wt% as Enclomiphene of formula (II) (0.466 mol). Then 0.24 L of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (100.8 g, 0.475 mol, 1.02 equiv.) was dissolved in absolute ethanol (1.7 L) and water (0.3 L), the solution was heated to 65°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 65°C. The dosage takes place in 30- 40 minutes. The inner temperature was decreased very slowly to 60°C over 80 minutes, then it was further decrease to 55°C over 40 minutes. When the inner temperature was in the range 60-55°C (typically at 58°C), the crystallization mixture was seeded with Enclomiphene citrate needle- shaped and a white product began to precipitate. Once reached 55°C the temperature was further decreased to 30°C over 30 minutes, then to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 0.4 L of absolute ethanol. The product was dried under vacuum at 65°C. At the end of drying, 269 g of Enclomiphene citrate of formula (I) as needle crystal were isolated, corresponding to 91.8% molar yield.

[00205] HPLC Analysis (A/A%): 99.79% Enchlomiphene, 0.04% Z-Clomiphene (i.e. Z-isomer).

[00206] Example 4: Preparation of Enclomiphene citrate of formula (I), having a needle shaped crystal habit, with a mixture of ethanol and water, wherein the amount of water is 15%.

(I)

[00207] Into a proper 1 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene of fomula (II) (15,0 g, assay 99.9 wt% 0.0369 mol, 1 equiv.) was dissolved in absolute ethanol (102 ml_, 6.8 mL/g of free base), then 18 ml_ (1.2 mL/g of free base) of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (7.92 g, 0.0377 mol, 1.02 equiv.) was dissolved in absolute ethanol (127 ml_) and water (23 ml_), the solution was heated to 65°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 65°C. The dosage takes place in 30-40 minutes. The inner temperature was decreased very slowly to 60°C over 80 minutes, then it was further decrease to 55°C over 40 minutes. When the inner temperature was in the range 60-55°C (typically at 58°C), the crystallization mixture was seeded with Enclomiphene citrate needle-shaped and a white product began to precipitate. Once reached 55°C the temperature was further decreased to 30°C over 30 minutes, then to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 30 ml_ of absolute ethanol. The product was dried under

vacuum at 65°C. At the end of drying, 20.2 g of Enclomiphene citrate of formula (I) as needle crystal were isolated, corresponding to 91.4% molar yield.

[00208] HPLC Analysis (A/A%): 99.86% Enchlomiphene, 0.03% Z-Clomiphene.

[00209] Example 4a: Preparation of Enclomiphene citrate of formula (I), having a needle shaped crystal habit, with a mixture of isopropanol and water, wherein the amount of water is 15%.

[00210] Into a proper 1 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene of fomula (II) (40,0 g, assay 99.9 wt% 0.0985 mol, 1 equiv.) was dissolved in isopropanol (272 ml_, 6.8 mL/g of free base), then 48 ml_ (1.2 mL/g of free base) of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (21.10 g, 0.100 mol, 1.02 equiv.) was dissolved in isopropanol (340 ml_, 8.5 mL/g of free base) and water (60 mL, 1.5 mL/g of free base), the solution was heated to 65°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 65°C. The dosage takes place in 30- 40 minutes. The inner temperature was decreased very slowly to 60°C over 80 minutes, then it was further decrease to 55°C over 40 minutes. When the inner temperature was in the range 60-55°C (typically at 58°C), the crystallization mixture was seeded with Enclomiphene citrate needle- shaped and a white product began to precipitate. Once reached 55°C the temperature was further decreased to 30°C over 30 minutes, then to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 30 mL of isopropanol. The product was dried under vacuum at 65°C. At the end of drying, 56.5 g of Enclomiphene citrate of formula (I) as needle crystal were isolated, corresponding to 95.9% molar yield.

[0021 1] Example 4b: Preparation of Enclomiphene citrate of formula (I), having a needle shaped crystal habit, with a mixture of n-propanol and water, wherein the amount of water is 15%.

[00212] Into a proper 0.5 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene of fomula (II) (9,0 g, assay 99.9 wt% 0.0985 mol, 1 equiv.) was dissolved in 7-propanol (61 mL, 6.8 mL/g of free base), then 1 1 ml_ (1.2 mL/g of free base) of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (4.70 g, 0.0224 mol, 1.02 equiv.) was dissolved in 7-propanol (77 ml_, 8.5 mL/g of free base) and water (14 ml_, 1.5 mL/g of free base), the solution was heated to 65°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 65°C. The dosage takes place in 30- 40 minutes. The inner temperature was decreased very slowly to 60°C over 80 minutes, then it was further decrease to 55°C over 40 minutes. When the inner temperature was in the range 60-55°C (typically at 58°C), the crystallization mixture was seeded with Enclomiphene citrate needle- shaped and a white product began to precipitate. Once reached 55°C the temperature was further decreased to 30°C over 30 minutes, then to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 30 mL of 7-propanol I. The product was dried under vacuum at 65°C. At the end of drying, 1 1.7 g of Enclomiphene citrate of formula (I) as needle crystal were isolated, corresponding to 88.1 % molar yield

[00213] Example 4c: Preparation of Enclomiphene citrate of formula (I), having a needle shaped crystal habit, with a mixture of n-butanol and water, wherein the amount of water is 15%.

[00214] Into a proper 0.5 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene of fomula (II) (9,0 g, assay 99.9 wt% 0.0985 mol, 1 equiv.) was dissolved in 7-butanol (61 mL, 6.8 mL/g of free base), then 1 1 mL (1.2 mL/g of free base) of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (4.70 g, 0.0224 mol, 1.02 equiv.) was dissolved in 7-butanol (77 mL, 8.5 mL/g of free base) and water (14 mL, 1.5 mL/g of free base), the solution was heated to 65°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 65°C. The dosage takes place in 30- 40 minutes. The inner temperature was decreased very slowly to 60°C over 80 minutes, then it was further decrease to 55°C over 40 minutes. When the inner temperature was in the range 60-55°C (typically at 58°C), the crystallization mixture was seeded with Enclomiphene citrate needle- shaped and a white product began to precipitate. Once reached 55°C the temperature was further decreased to 30°C over 30 minutes, then to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 30 ml_ of 7-butanol. The product was dried under vacuum at 65°C. At the end of drying, 1 1.6 g of Enclomiphene citrate of formula (I) as needle crystal were isolated, corresponding to 87.4% molar yield.

[00215] Example 4d: Preparation of Enclomiphene citrate of formula (I), having a needle shaped crystal habit, with a mixture of tert-butanol and water, wherein the amount of water is 15%.

[00216] Into a proper 0.5 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene of fomula (II) (9,0 g, assay 99.9 wt% 0.0985 mol, 1 equiv.) was dissolved in te T-butanol (61 ml_, 6.8 mL/g of free base), then 1 1 ml_ (1.2 mL/g of free base) of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (4.70 g, 0.0224 mol, 1.02 equiv.) was dissolved in te T-butanol (77 ml_, 8.5 mL/g of free base) and water (14 mL, 1.5 mL/g of free base), the solution was heated to 65°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 65°C. The dosage takes place in 30- 40 minutes. The inner temperature was decreased very slowly to 60°C over 80 minutes, then it was further decrease to 55°C over 40 minutes. When the inner temperature was in the range 60-55°C (typically at 58°C), the crystallization mixture was seeded with Enclomiphene citrate needle- shaped and a white product began to precipitate. Once reached 55°C the temperature was further decreased to 30°C over 30 minutes, then to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 30 mL of te T-butanol. The product was dried under vacuum at 65°C. At the end of drying, 1 1.2 g of Enclomiphene citrate of formula (I) as needle crystal were isolated, corresponding to 84.4% molar yield.

[00217] Example 5: Preparation of Enclomiphene citrate of formula (I), having a needle shaped crystal habit. Preparation of the seed crystal.

[00218] Into a proper 1 L reactor, equipped with propeller, temperature probes, condenser; Enclomiphene of fomula (II) (15,0 g, assay 99.9 wt% 0.0369 mol, 1 equiv.) was dissolved in absolute ethanol (102 ml_, 6.8 mL/g of free base), then 18 ml_ (1.2 mL/g of free base) of water were added and the solution was heated to 65°C. Meanwhile, citric acid monohydrate (7.92 g,

0.0377 mol, 1.02 equiv.) was dissolved in absolute ethanol (127 ml_, 8.5 mL/g of free base) and water (23 mL 1.5 mL/g of free base), the solution was heated to 50°C. The solution of citric acid was dropped into the solution of Enclomiphene (II), while maintaining 50°C. The dosage takes place in 30-40 minutes. At the end of the dosage, the stirring was turned off and the mixture was allowed to cool down to room temperature without stirring. The product began to crystallize at 40-30°C. Once reached 20- 25°C the stirring was turned on and the temperature was further decreased to 0°C over 30 minutes. The slurry was stirred at 0°C for at least two hours, then it was filtered and the wet cake was washed with 30 mL of absolute ethanol. The product was dried under vacuum at 65°C. At the end of drying, 13.9 g of Enclomiphene citrate of formula (I) were isolated, corresponding to 62.3% molar yield

[00219] Example 6: Preparation of Enclomiphene citrate of formula (I), having a non-needle shaped crystals, with a mixture of acetone and water, wherein the amount of water is 15%.

Comparative example (see Fig. 8) and evidence example of the invention. Following the same process described in the example 4, substituting ethanol solvent with acetone solvent. Starting from 15,0 g of Enclomiphene of formula (II), following the above mentioned process, 22.3 g of Enclomiphene citrate of formula (I) were isolated, corresponding to 94.2% molar yield product. For the morphology of the crystal see fig. 8.

[00220] Indeed, the microscopy analysis provides a better further evidence of the crystal habit of Enclomiphene citrate (I) of the example 6 (see Fig.8) which has a form more different than/to Enclomiphene citrate (I) having a needle shaped crystal habit, obtained according to above described examples,

1. e. 4, 4a, 4b, 4c, 4d (see Fig. 5, 6 and 7).

[00221] HPLC Analysis (A/A%): 99.63% Enchlomiphene, 0.20% Z-Clomiphene.

[00222] Example 7: Analytical method to identify and quantify Z-Clomiphene of formula (IV) into Enclomiphene of formula (II) or Enclomiphene citrate of formula (I) or Enclomiphene BPA salt of formula (III) and for determining the chemical purity.

[00223] Chromatographic conditions:

Dim. Column: 250 mm x 4.6 mm , 5 pm

Stationaly phase: Butyl sylane (USP phase L26, Vydac 4C is suggested) Temp. Column: room temperature

Mobile Phase: Methanol / water / triethylamine 55 : 45 : 0.3 v/v

Adjust at pH 2.5 with phosphoric acid

Flow: 1.0 mL/min

Detector UV a 233 nm,

Injection Volume: 10 μΙ_

Sample diluent: mobile phase.

Applying the conditions described above the expected retention times are as indicated below:

/////////////////Enclomiphene citrate, New patent, WO 2017182097, F.I.S. – FABBRICA ITALIANA SINTETICI S.P.A

File:Enclomiphene.png

Enclomiphene

Synonyms: Chloramiphene Citrate; Citrato de cloramifeno; Clomifencitrat; Clomifène, citrate de; Clomifeni Citras; Clomifeno, citrato de; Clomiphene Citrate; Klomifeenisitraatti; Klomifen Sitrat; Klomifen-citrát; Klomifén-citrát; Klomifencitrat; Klomifeno citratas; MER-41; MRL-41; NSC-35770; クロミフェンクエン酸塩
BAN: Clomifene Citrate [BANM]
USAN: Clomiphene Citrate
INN: Clomifene Citrate [rINNM (en)]
INN: Citrato de clomifeno [rINNM (es)]
INN: Clomifène, Citrate de [rINNM (fr)]
INN: Clomifeni Citras [rINNM (la)]
INN: Кломифена Цитрат [rINNM (ru)]
Chemical name: A mixture of the E and isomers of 2-[4-(2-chloro-1,2-diphenylethenyl)phenoxy]-N,N-diethylethanamine dihydrogen citrate
Molecular formula: C26H28ClNO,C6H8O7 =598.1
CAS: 911-45-5 (clomifene);15690-57-0((E)-clomifene ); 15690-55-8 ((Z)-clomifene); 50-41-9 (clomifene citrate); 7599-79-3 ((E)-clomifene
citrate); 7619-53-6 ((Z)-clomifene citrate)
ATC code: G03GB02
ATC code (veterinary): QG03GB02
UNII code: 1B8447E7YI (clomifene citrate); UY5X264QZV ((Z)-clomifene citrate)

Chemical Structure of ClomifeneChemical Structure of Clomifene

NOTE:

Clomifene may be separated into its Z-and E-isomers, zuclomifene and enclomifene
.


Filed under: PATENT, PATENTS Tagged: Enclomiphene citrate, F.I.S. – FABBRICA ITALIANA SINTETICI S.P.A, NEW PATENT, WO 2017182097

FDA approves new treatment for adults with mantle cell lymphoma

$
0
0

FDA approves new treatment for adults with mantle cell lymphoma

The U.S. Food and Drug Administration today granted accelerated approval to Calquence (acalabrutinib) for the treatment of adults with mantle cell lymphoma who have received at least one prior therapy.

“Mantle cell lymphoma is a particularly aggressive cancer,” said Richard Pazdur, M.D., director of the FDA’s Oncology Center of Excellence and acting director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research. “For patients who have not responded to treatment or have relapsed, Calquence provides a new treatment option that has shown high rates of response for some patients in initial studies.” Continue reading.


Filed under: FDA 2017 Tagged: acalabrutinib, Calquence, FDA 2017

GLIMEPIRIDE

$
0
0

ChemSpider 2D Image | Glimepiride | C24H34N4O5S

glimepiride.png

Glimepiride.svg

Glimepiride

  • Molecular FormulaC24H34N4O5S
  • Average mass490.616 Da
  • HOE 490
    UNII:6KY687524K
3-Ethyl-N-{2-[4-({(E)-hydroxy[(trans-4-methylcyclohexyl)imino]methyl}sulfamoyl)phenyl]ethyl}-4-methyl-2-oxo-2,5-dihydro-1H-pyrrole-1-carboximidic acid
93479-97-1 [RN]
1-{[4-(2-{[(3-ethyl-4-methyl-2-oxo-2,5-dihydro-1H-pyrrol-1-yl)carbonyl]amino}ethyl)phenyl]sulfonyl}-3-(trans-4-methylcyclohexyl)urea
3-Ethyl-4-methyl-N-[2-(4-{[(trans-4-methylcyclohexyl)carbamoyl]sulfamoyl}phenyl)ethyl]-2-oxo-2,5-dihydro-1H-pyrrole-1-carboxamide
1H-Pyrrole-1-carboxamide, 3-ethyl-2,5-dihydro-4-methyl-N-[2-[4-[[[[(trans-4-methylcyclohexyl)amino]carbonyl]amino]sulfonyl]phenyl]ethyl]-2-oxo-
Amarel [Trade name]
Amaryl [Trade name]
Endial [Trade name]

Glimepiride (original trade name Amaryl) is an orally available medium-to-long-acting sulfonylurea antidiabetic drug. It is sometimes classified as either the first third-generation sulfonylurea,[1] or as second-generation.[2]

Glimepiride is a Sulfonylurea. The chemical classification of glimepiride is Sulfonylurea Compounds.

Glimepiride is a long-acting, third-generation sulfonylurea with hypoglycemic activity. Compared to other generations of sulfonylurea compounds, glimepiride is very potent and has a longer duration of action. This agent is metabolized by CYP2C9 and shows peroxisome proliferator-activated receptor gamma (PPARgamma) agonistic activity.

Glimepiride is only found in individuals that have used or taken this drug. It is the first III generation sulphonyl urea it is a very potent sulphonyl urea with long duration of action. The mechanism of action of glimepiride in lowering blood glucose appears to be dependent on stimulating the release of insulin from functioning pancreatic beta cells, and increasing sensitivity of peripheral tissues to insulin. Glimepiride likely binds to ATP-sensitive potassium channel receptors on the pancreatic cell surface, reducing potassium conductance and causing depolarization of the membrane. Membrane depolarization stimulates calcium ion influx through voltage-sensitive calcium channels. This increase in intracellular calcium ion concentration induces the secretion of insulin.

Indications

Glimepiride is indicated to treat type 2 diabetes mellitus; its mode of action is to increase insulin production by the pancreas. It is not used for type 1 diabetes because in type 1 diabetes the pancreas is not able to produce insulin.[3]

Contraindications

Its use is contraindicated in patients with hypersensitivity to glimepiride or other sulfonylureas.

Adverse effects

Side effects from taking glimepiride include gastrointestinal tract (GI) disturbances, occasional allergic reactions, and rarely blood production disorders including thrombocytopenialeukopenia, and hemolytic anemia. In the initial weeks of treatment, the risk of hypoglycemia may be increased. Alcohol consumption and exposure to sunlight should be restricted because they can worsen side effects.[3]

Pharmacokinetics

Two generic oral tablets of glimepiride, 2 mg each

Gastrointestinal absorption is complete, with no interference from meals. Significant absorption can occur within one hour, and distribution is throughout the body, 99.5% bound to plasma protein. Metabolism is by oxidative biotransformation, it is hepatic and complete. First, the medication is metabolized to M1 metabolite by CYP2C9. M1possesses about ​13 of pharmacological activity of glimepiride, yet it is unknown if this results in clinically meaningful effect on blood glucose. M1 is further metabolized to M2metabolite by cytosolic enzymes. M2 is pharmacologically inactive. Excretion in the urine is about 65%, and the remainder is excreted in the feces.

Mechanism of action

Like all sulfonylureas, glimepiride acts as an insulin secretagogue.[4] It lowers blood sugar by stimulating the release of insulin by pancreatic beta cells and by inducing increased activity of intracellular insulin receptors.

Not all secondary sufonylureas have the same risks of hypoglycemia. Glibenclamide (glyburide) is associated with an incidence of hypoglycemia of up to 20–30%, compared to as low as 2% to 4% with glimepiride. Glibenclamide also interferes with the normal homeostatic suppression of insulin secretion in reaction to hypoglycemia, whereas glimepiride does not. Also, glibenclamide diminishes glucagon secretion in reaction to hypoglycemia, whereas glimepiride does not.[5]

Image result for SYNTHESIS Glimepiride

Image result for SYNTHESIS Glimepiride

Interactions

Nonsteroidal anti-inflammatory drugs (such as salicylates), sulfonamideschloramphenicolcoumadin and probenecid may potentiate the hypoglycemic action of glimepiride. Thiazides, other diuretics, phothiazides, thyroid products, oral contraceptives, and phenytoin tend to produce hyperglycemia.

 

SYNTHESIS

EP 0031058; US 4379785, Arzneim-Forsch Drug Res 1988,38(8),1079

The condensation of 3-ethyl-4-methyl-3-pyrrolin-2-one (I) with 2-phenylethyl isocyanate (II) at 150 C gives 3-ethyl-4-methyl-2-oxo-N-(2-phenylethyl)-3-pyrrolin-1-carboxamide (III), which is sulfonated with chlorosulfonic acid at 40 C to yield the corresponding benzenesulfonyl chloride (IV). The reaction of (IV) with concentrated NH4OH affords the sulfonamide (V), which is finally condensed with 4-methylcyclohexyl isocyanate (VI) in acetone.

clip

Image result for SYNTHESIS Glimepiride

CLIP

Image result for SYNTHESIS Glimepiride

Following is one of the synthesis routes: 3-Ethyl-4-methyl-3-pyrrolin-2-one could be condensed (I) with 2-phenylethyl isocyanate (II) at 150 C to produce 3-ethyl-4-methyl-2-oxo-N-(2-phenylethyl)-3-pyrrolin-1-carboxamide (III), which is sulfonated with chlorosulfonic acid at 40 C to yield the corresponding benzenesulfonyl chloride (IV). The reaction of (IV) with concentrated NH4OH affords the sulfonamide (V), which is finally condensed with 4-methylcyclohexyl isocyanate (VI) in acetone.

CLIP

http://science24.com/paper/6906

Image result for SYNTHESIS Glimepiride

PAPER

https://www.sciencedirect.com/science/article/pii/S073170850500378X

Image result for SYNTHESIS Glimepiride

PATENT

https://www.google.com/patents/US20070082943

  • Glimepiride, according to U.S. Pat. No. 4,379,785 (EP 031058) issued to Hoechst is prepared via reaction of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (IV) with trans-4-methylcyclohexyl isocyanate (VIII). U.S. Pat. No. 4,379,785 (EP 031058) (hereafter referred to as the ‘785 patent) discloses heterocyclic substituted sulfonyl ureas, particularly 3-Ethyl-2,5-dihydro-4-methyl-N-[2-[4-[[[[(trans-4-methyl cyclohexyl)amino]carbonyl]amino]sulfonyl]phenyl]ethyl]-2-oxo-1H-pyrrole-1-carboxamide i.e. Glimepiride (I). The ‘785 patent teaches the preparation of Glimepiride starting from 3-Ethyl-4-methyl-3-pyrolidine-2-one (II) and 2-phenyl ethyl isocyanate to give [2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene (III). The [2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene is converted to the 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (IV), by reacting with chlorosulphonic acid, followed by treatment with ammonia solution. This intermediate compound (IV) is then finally reacted with trans-4-methylcyclohexyl isocyanate (VIII) prepared from trans-4-methyl cyclohexylamine HCl (VII) to form Glimepiride.
  • [0004]
    Glimepiride can also be synthesized by reaction of N-[[4-[2-(3-ethyl-4-methyl-2-oxo-3-pyrroline-1-carboxamido)-ethyl]phenyl]sulphonyl]methylurethane (IX) with trans-4-methyl cyclohexyl amine (VII) as reported by R. Weyer, V. Hitzel in Arzneimittel Forsch 38, 1079 (1988).
  • [0005]
    trans-4Methylcyclohexyl isocyanate (VIII) is prepared from trans-4-methyl cyclohexyl amine HCl (VII), by phosgenation.
  • [0006]
    H. Ueda et. al., S.T.P Pharma Sciences, 13(4) 281-286, 2003 discloses a novel polymorph of Glimepiride, Form II obtained by recrystallisation from a solvent mixture of ethanol and water. It also discloses that earlier known form is Form I. Reported solvents for obtaining Form I are methanol, acetonitrile, chloroform, butyl acetate, benzene and toluene.
  • [0007]
    An alternative route is disclosed in WO03057131(Sun Pharmaceutical), where 3-ethyl-4-methyl-2,5-dihydro-N-(4-nitrophenyloxycarbonyl)-pyrrole-2-one is treated with 4-(2-aminoethyl)-benzene sulphonamide to obtain 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (IV) which was then converted to Glimepiride (I). However, nonavailability of raw material and the yield being poor, the process as described in U.S. Pat. No. 4,379,785 is preferred.
  • [0008]
    To obtain Glimepiride of highest purity, following intermediates should be of highest quality:
  • [0009]
    a) 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (IV) with lowest possible content of ortho and meta isomers.
  • [0010]
    b) Trans-4-methyl cyclohexyl amine (VII) and its respective isocyanate (VIII) should have lowest content of the cis isomer.
  • [0011]
    The preparation of the 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide is well disclosed in the patent U.S. Pat. No. 4,379,785. It is prepared by condensation of 3-ethyl-4-methyl-3-pyrrolidine-2-one of Formula (II) with 2-phenyl ethyl isocyanate. The condensed product is then chlorosulphonated with chlorosulphonic acid followed by ammonolysis with liq. ammonia to give compound of Formula (IV). The purity is not well documented in the patents, and by following the patented process, ˜85 to 88% of desired para isomer is obtained. This is evident as the chlorosulphonation is ortho-para directing.
  • [0012]
    Hence, there is a need to develop purification process to maintain undesired ortho and meta isomers below 0.1%.
  • [0013]
    The other key intermediate trans-4-methylcyclohexyl amine HCl (VII) should preferably have lowest possible content of the cis isomer. The commonly used procedure is reduction of 4-methyl cyclohexanone oxime (V) with sodium in alcohol, preferably ethanol.
  • [0014]
    T. P. Johnston, et. al., J. Med. Chem., 14, 600-614 (1971); H. Booth, et. al., J. Chem. Soc (B) 1971, 1047-1050 and K. Ramalingam et. al., Indian Journal of Chem Vol. 40, 366-369 (April 1972) all report the abovementioned reduction. The amine obtained via this process typically contains between 8 to 10% of the cis isomer. However, use of high excess sodium metal (25 eqv.) for reduction makes process commercially and environmentally unviable. Also, the purification of trans amine from the mixture via the distillation is very difficult as the boiling points differ only by about 2° C. Also there is an inherent drawback of said free amine as, it immediately forms carbonate salt. Further purification of the amine to reduce the cis content via crystallization of its salt is not sufficiently documented. Prior art describes purification of crude trans-4-methylcyclohexylamine HCl by crystallization of its hydrochloride but the yield and purity are not sufficiently discussed. A description of such purification is provided in J. Med. Chem, 14, 600-614 (1971), wherein trans-4-methylcyclohexylamine HCl is obtained by triple crystallization in acetonitrile of the crude hydrochloride (m.p. 260° C.) in 27% yield.
  • [0015]
    WO 2004073585 (Zentiva) describes a process for preparation of trans-4-methylcyclohexylamine HCl wherein the highlights of the invention are the use of sodium metal and purification via the pivalic acid salt. However drawbacks of the process are use of sodium metal, which is hazardous and pivalic acid which is expensive. The overall yield is ˜40%.
  • [0016]
    Thus considering the current stringent pharmacopieal requirements for cis content, there is a need for obtaining Glimepiride having cis impurity content well below 0.15% by a cost effective process.
  • [0017]
    Key factors in the production of Glimepiride are:
  • [0018]
    a) Substantial purity of trans-4-methyl cyclohexyl amine HCl (VII) with the lowest possible content of the cis isomer.
  • [0019]
    b) Substantial purity of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (IV) with the lowest possible content of the ortho and meta isomer.
  • [0020]
    The purity of intermediate compound of Formula (IV) when prepared by the process disclosed in ‘785 patent, was found to be 82 to 85% by HPLC.
    • schemes I to III.

      Figure US20070082943A1-20070412-C00001
      Figure US20070082943A1-20070412-C00002
      Figure US20070082943A1-20070412-C00003
    • [0037]
      The purification of trans-4-methyl cyclohexylamine HCl (VII) is accomplished by using an appropriate solvent combination. The mixture of cis/ trans stereoisomers (i.e. 50:50) were dissolved in diluted methanol and the desired trans isomer is coprecipitated by adding acetone to it. The process is repeated with different proportions of the solvent mixture to get the trans-4-Methyl cyclohexylamine HCl (VII) >99.5% with cis isomer less than 0.15%. The overall yield from 4-methyl cyclohexanone is ˜30%. The purification has been achieved using a solvent mixture of alcohol and ketone. A preferred alcohol for dissolution is an aliphatic one wherein carbon chain may be preferably C1-C4. Preferably methanol is used to dissolve the crude trans-4-Methyl cyclohexylamine HCl. The ratio of substrate:methanol:acetone is fixed at 1:1.5:6 for achieving the desired purity. The cosolvent used for precipitation is an aliphatic ketone. The preferred ketone is acetone. The precipitation is carried out at a temperature between 20 to 50° C., preferably between 30 to 50° C. and most preferably at about 40° C. The addition of acetone is carried out over a period of 2 to 6 hrs, more preferably for about 2 to 4 hrs and most preferably in about 3 hrs. The compound thus obtained has a purity >95% by gas chromatography.
    • [0038]
      The enriched trans-4-Methyl cyclohexylamine HCl (VII) (>95%) is further purified using different proportions of the same solvent mixture. The enriched trans isomer is dissolved in alcohol and reprecipitated using an aliphatic ketone. The ratio of substrate:methanol:acetone ratio is fixed at 1:1.5:13.6 for obtaining purity greater than 99.8%.

Image result for SYNTHESIS Glimepiride

    • EXAMPLE 1trans-4-Methyl cyclohexylamine HCl (VII)
    • [0053]
      1.5 Kg of crude 4-Methyl cyclohexyloxime (V) was dissolved in 8.33 L Methanol. To this 0.15 Kg Raney nickel was added. Then the mixture was hydrogenated at 4-5 Kg/cmpressure at 50 to 55° C. After the absorption of Hceases, the reaction mass is cooled down and filtered. From resulting reaction mixture, methanol was distilled completely. Crude concentrated oil obtained is cooled to 15 to 20° C. to which methanolic hydrochloric acid (12 to 13%) is added slowly, when the product i.e. 4-Methylcyclohexylamine HCl precipitates out. The yield obtained 1.5 Kg of crude 4-methyl cyclohexylamine HCl (85%) with ˜50% content of trans isomer. The crude 4-Methyl cyclohexylamine HCl 1.5 Kg (wet) was further purified in methanol/acetone mixture. The crude 4-methyl cyclohexylamine HCl (1.5 Kg) was dissolved in 2.25 L of methanol at 25 to 30° C. Slowly started addition of 13.5 L of acetone over a period of 3 hrs. The trans-4-methyl cyclohexylamine HCl precipitated out. Yield 0.6 Kg. The purity achieved of trans isomer is >95%. The cis isomer at this stage is ˜2 to 3%.
    • [0054]
      The trans-4-methyl cyclohexylamine HCl (0.6 Kg) thus obtained is again taken in 0.9 L of methanol and is dissolved completely at 25 to 30° C. 8.1 L acetone is added slowly over a period of 3 hrs when pure trans isomer precipitates out completely. The purity achieved at this stage is >99.8% and cis isomer well below 0.15%. The yield thus obtained after the second purification is 0.48 Kg of trans-4-Methyl cyclohexylamine HCl (27.2% yield calculated on the starting oxime). Purity of the desired trans isomer is greater than 99.8% by G.C.
    • [0055]
      Melting point of the trans-4-methyl cyclohexylamine HCl thus obtained is 262° C. to 263° C.

EXAMPLE 2Preparation of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (IV)

    • [0056]
      3-Ethyl-4-methyl-2,5-dihydro-1H-pyrrole-2-one (II) (1.0 Kg) and β-phenylethyl isocyanate (1.488 Kg) were mixed in anhydrous toluene (4.0 L) and refluxed for 4 hrs. The toluene was distilled off and hexane (8.0 L) was added to the reaction mixture at 50° C. The product precipitated is cooled to 0 to 5° C. to obtain the solid compound viz. 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene (2.17 Kg). It was filtered & washed with 2.0 L of hexane.
    • [0057]
      To a cooled (15 to 25° C.) solution of chlorosulfonic acid (2.8 L), 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene (2.0 Kg) was added in small portions over a period of 2 to 3 hrs. Further it was stirred for 30 min at this temperature and then temperature was gradually raised to 30 to 35° C. The reaction mass is stirred further for 2 hrs. The reaction mixture was then quenched into ice-water and stirred for 1 hr and filtered to obtain the product 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonyl chloride (2.0 kg). To a cooled (15 to 20° C.) solution of diluted ammonia (1.4 L) 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonyl chloride was added in small portion over 1 to 2 hrs. The reaction mixture was then heated to 70° C. for 2 hrs when ammonolysis is complete. The product converted is then stirred for 1 hr at R.T. and filtered and dried at 90 to 100° C. to obtain crude 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (2.2 Kg) having HPLC purity in the range of 82 to 88%. The crude compound 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (2.2 Kg) is then purified from mixture of organic solvents chosen from Methanol, Acetone & toluene.

EXAMPLE 3APurification of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonamide (IV)

    • [0058]
      1st Purification
    • [0059]
      In a reaction vessel containing Toluene (12.0 L), 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide (2.0 Kg) was charged at 25 to 30° C. Slowly the temperature was raised to 60 to 65° C. and methanol (5.0 L) was added via the dosing tank slowly when the product dissolved completely. Refluxed it for 0.5 hr. Charcoalised and filtered the product in another reaction vessel. Distill off toluene/methanol mixture till total recovery about 65% under vacuum. White crystalline product precipitated out. After the recovery, cool the reaction mass to 15 to 20° C. The resulting crystallized solid product was filtered and washed two times with chilled acetone (about 2 L) each. The resulting product was dried at 90 to 100° C. in air oven till constant weight to obtain about 1.4 Kg of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonamide with greater than 95% HPLC purity.

EXAMPLE 3BPurification of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonamide (IV)

    • [0060]
      2nd Purification
    • [0061]
      In a reaction vessel containing Acetone (8.4 L), (1.4 Kg) of 1st purified 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonamide was charged at 25 to 30° C. slowly and the temperature was raised to 55 to 60° C. Methanol was added (5.6 L) via the dose tank at this reflux temperature to dissolve it completely. Refluxed it for further 30 min. Distilled off acetone/ methanol mixture till total recovery about 65 to 70%. White crystalline product precipitated out. After the recovery slowly cooled the product to 15 to 20° C. The resultant solid product was filtered, washed two times with chilled acetone (1.4 L) each. The 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido) ethyl] benzene sulfonamide was dried at 90 to 100° C. in air oven till constant weight to obtain about 1.12 Kg of 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonamide (IV) with greater than 99.5% purity with other isomers i.e. ortho and meta well below 0.2% respectively.

EXAMPLE 4Preparation of 3-Ethyl-2,5-dihydro-4-methyl-N-[2-[4-[[[[(trans-4-methyl cyclohexyl)amino]carbonyl]amino]sulfonyl]phenyl]ethyl]-2-oxo-1 H-pyrrole-1-carboxamide (I).

    • [0062]
      In a reaction vessel containing (24.2 L) Acetone, 4-[2-(3-Ethyl-4-methyl-2-carbonyl pyrrolidine amido)ethyl] benzene sulfonamide (1.0 Kg) and potassium carbonate (0.46 Kg) was added and refluxed at about 55 to 60° C. for 1 hr. trans-4-Methyl-cyclohexyl isocyanate was obtained by method known in art from trans-4-methyl-cyclohexylamine. A solution of trans-4-methyl-cyclohexyl isocyanate (0.515 Kg) in toluene (5 L) was prepared and added to the above reaction mixture. This reaction mixture is refluxed for 12 hrs, then cooled. To this cooled reaction mass charge 27 L of water. The reaction mass was filtered and the pH was adjusted to 5.5 to 6.0 by adding acetic acid at about 20 to 25° C. The solid obtained was filtered and washed with water. The 3-Ethyl-2,5-dihydro-4-methyl-N-[2-[4-[[[[(trans-4-methyl cyclohexyl)amino]carbonyl]amino]sulfonyl]phenyl]ethyl]-2-oxo-1H-pyrrole-1-carboxamide (I) obtained is then dried at 90 to 100° C. till constant weight. Yield of the product is 86.3%.

EXAMPLE 5Purification of 3-Ethyl-2,5-dihydro-4-methyl-N-[2-[4-[[[[(trans-4-methyl cyclohexyl)amino]carbonyl]amino]sulfonyl]phenyl]ethyl]-2-oxo-1H-pyrrole-1-carboxamide (I)

  • [0063]
    In a reaction vessel containing 6.0 L methanol and 1.0 Kg crude Glimepiride, dry ammonia gas was purged at 20 to 25° C. till all Glimepiride dissolves and a clear solution is obtained. This homogeneous mass was then charcoalised, filtered and finally neutralized with Glacial acetic acid to pH 5.5 to 6.0, till the entire product precipitates out. The pure Glimepiride was then filtered and dried at 65° C. to 70° C. till constant weight. Yield obtained was ˜90%.

CLIP

  • Journal of Pharmaceutical Sciences 100(11):4700-9
  • DOI
  • 10.1002/jps.22662

Image result for GLIMEPIRIDE NMR

Magnified 1H NMR spectra of (a) glimepiride and its solid dispersions with hyperbranched polymers containing the (b) hydroxyl and (c) the tertiary amino functional groups.

Magnified 13C NMR spectra of (a) glimepiride and its solid dispersions with hyperbranched polymers containing (b) the hydroxyl and (c) the tertiary amino functional groups.

The difference spectra of the solid dispersions of glimepiride and the hyperbranched polymer containing (a) the hydroxyl groups and (b) the tertiary amino groups. The difference spectra were obtained by subtraction of the spectra of the pure hyperbranched polymers from the spectra of the solid dispersions. The ATR spectra of the pure hyperbranched polymers were recorded on samples that were prepared under the same conditions as solid dispersions, only without the presence of the glimepiride drug.

Patents

  1. US6150383
  2. US6211205
  3. US6303640
  4. US6329404
  5. US8071130
  6. US7538125
  7. US7700128
  8. US7358366

FDA Orange Book Patents

FDA Orange Book Patents: 1 of 3 (FDA Orange Book Patent ID)
Patent 7358366
Expiration Apr 19, 2020. 7358366*PED expiration date: Oct 19, 2020
Applicant SB PHARMCO
Drug Application
  1. N021700 (Prescription Drug: AVANDARYL. Ingredients: GLIMEPIRIDE
  2. ROSIGLITAZONE MALEATE)
FDA Orange Book Patents: 2 of 3 (FDA Orange Book Patent ID)
Patent 8071130
Expiration Jun 8, 2028
Applicant TAKEDA PHARMS USA
Drug Application
  1. N021925 (Prescription Drug: DUETACT. Ingredients: GLIMEPIRIDE
  2. PIOGLITAZONE HYDROCHLORIDE)
  3. N021925 (Prescription Drug: DUETACT. Ingredients: GLIMEPIRIDE
  4. PIOGLITAZONE HYDROCHLORIDE)
FDA Orange Book Patents: 3 of 3 (FDA Orange Book Patent ID)
Patent 7700128
Expiration Jan 30, 2027
Applicant TAKEDA PHARMS USA
Drug Application
  1. N021925 (Prescription Drug: DUETACT. Ingredients: GLIMEPIRIDE
  2. PIOGLITAZONE HYDROCHLORIDE)
  3. N021925 (Prescription Drug: DUETACT. Ingredients: GLIMEPIRIDE
  4. PIOGLITAZONE HYDROCHLORIDE)

CLIP

STR2

Journal of  China Pharmaceutical University       1999 , 30(3):163 ~ 165

Ethyl acetoacetate (2) Preparation of literature more, such as with ethyl iodide or ethyl bromide as ethyl reagents will produce a double ethylation or oxyethylation, it is difficult to separate. We use dimethylamine and ethyl acetoacetate reaction enamine, then diethyl sulfate as ethylating agent, you can reduce the side reactions, product purity, the yield up to 80%. Preparation of cyanohydrin (3) Hydrochloric acid anhydrous literature, toxicity, difficult to operate, we use solid sodium cyanide and sodium bisulfite in the aqueous phase reaction, get 3, easy to operate. In the literature 1-acetyl-3- Ethyl-4-methyl-3-pyrrolin-2-one (4) was purified by high vacuum distillation and then hydrolyzed to give 3-ethyl- In the distillation of the product easy to loss, after the change to the crude hydrolysis, two-step yield of 33%. Reported in the literature 5 and phenethyl isocyanate (6) without solvent direct reaction of 3-ethyl-4-methyl-2-oxo-3-pyrroline-1 – N- (2 – phenethyl) A Amide (7), the experiment found that the reaction heat when the heating easy to red material, we add toluene as a solvent, the reaction is smooth, easy to post-treatment. 6 preparation, the general method is to use phosgene, but phosgene often Temperature of gas, highly toxic, difficult to operate, we use triphosgene instead of triphosgene as a yellow solid, easy to transport, weighing, laboratory convenience. We refer to the process of domestic glyburide, 7 chlorosulfonated, ammoniated sulfonamide (9), two-step yield of 77%. The last 9 reacts with trans-4-methylcyclohexylisocyanate to form glimepiride (1). Ethyl acetoacetate as the starting material, eight-step total yield of 11.5%.

Journal of  China Pharmaceutical University       1999 , 30(3):163 ~ 165

Glimepi ride (1) trade name Amary l, chemical name 1- [4- [2- (3-ethyl-4-methyl-2-oxo-3-pyrroline-1-carboxamido ) Ethyl] phenylsulfonyl] -3- (trans-4-methylcyclohexyl) urea, a new sulfonylurea hypoglycemic agent developed by Hoechst AG in Germany and listed in the Netherlands and Switzerland in 1995, In 1996 the United States FDA approval

  • 1 Campbell RK.Glimepiride :Role  of  a  new   sulfonylurea  in  the t reatment of type 2  diabetes mellitus .An n Pharmacother ,  1998 , 32 :1044
  • 2 Hans P ,   Joachim  K .Synt hesis  of  oxyopsopy rrolecarboxyli c acid

and  further  investigations  in  the  pyrrolone  series.Ann  Chem ,

1964 , 680 :60

  • 3 Glimepiride.Dr ugs F ut , 1992 , 17(9):774
  • 4 Corson BB,   Dodge  RA ,   Harris  SA ,   et  al .M andeli c acid.Org Syn ,  1941 ,   Coll Vol 1 :329
  • 5 M aurice WG ,  Roy  VD,   Brian  I ,   et  a l .A new  synt hesis  of iso- cyanates .J  Chem Soc ,  Perkin  Ⅰ ,  1976 :141
  • 6 天津医药工业研究所.糖尿病药物-优降糖的新合成法.医药工业, 1974 , 4 :11
  • 7 Weyer R, Gei sen K , Hitzel V ,   et  al .Heterocyclic subst ituted sul- f onyl ureas and their  use .Ger O f fen ,  1979 :2951135 A 1
Cited Patent Filing date Publication date Applicant Title
US4379785 * Dec 17, 1980 Apr 12, 1983 Hoechst Aktiengesellschaft Heterocyclic substituted sulfonyl ureas, and their use

References

  1. Jump up^ Hamaguchi T, Hirose T, Asakawa H, et al. (December 2004). “Efficacy of glimepiride in type 2 diabetic patients treated with glibenclamide”. Diabetes Res. Clin. Pract. 66 Suppl 1: S129–32. doi:10.1016/j.diabres.2003.12.012PMID 15563963.
  2. Jump up^ Davis SN (2004). “The role of glimepiride in the effective management of Type 2 diabetes”. J. Diabetes Complicat18 (6): 367–76. doi:10.1016/j.jdiacomp.2004.07.001PMID 15531188.
  3. Jump up to:a b “Glimepiride: MedlinePlus Drug Information”nih.gov.
  4. Jump up^ Nissen SE, Nicholls SJ, Wolski K, et al. (April 2008). “Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial”. JAMA299 (13): 1561–73. doi:10.1001/jama.299.13.1561PMID 18378631.
  5. Jump up^ Davis, Stephen N. (2005). “60. Insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas”. In Brunton, Laurence L.; Lazo, John S.; Parker, Keith L. (eds.). Goodman & Gilman’s The Pharmacological Basis of Therapeutics. New York: McGraw-Hill. p. 1636. ISBN 0-07-142280-3.

External links

Title: Glimepiride
CAS Registry Number: 93479-97-1
CAS Name: 3-Ethyl-2,5-dihydro-4-methyl-N-[2-[4-[[[[(trans-4-methylcyclohexyl)amino]carbonyl]amino]sulfonyl]phenyl]ethyl]-2-oxo-1H-pyrrole-1-carboxamide
Additional Names: N-[4-[2-(3-ethyl-4-methyl-2-oxo-3-pyrroline-1-carboxamido)-ethyl]-benzenesulfonyl]-N¢-4-methylcyclohexylurea; 1-[4-[2-(3-ethyl-4-methyl-2-oxo-3-pyrroline-1-carboxamido)ethyl]phenylsulfonyl]-3-(4-methylcyclohexyl)urea
Manufacturers’ Codes: HOE-490
Trademarks: Amaryl (Aventis)
Molecular Formula: C24H34N4O5S
Molecular Weight: 490.62
Percent Composition: C 58.75%, H 6.99%, N 11.42%, O 16.31%, S 6.54%
Literature References: Sulfonylurea. Prepn: R. Weyer et al., DE 2951135eidem, US 4379785 (1981, 1983 both to Hoechst). Synthesis: R. Weyer, V. Hitzel, Arzneim.-Forsch. 38, 1079 (1988). Pharmacology: K. Geisen, ibid., 1120. Effects on insulin and glucagon secretion: V. Leclercq-Meyer et al., Biochem. Pharmacol. 42, 1634 (1991). HPLC determn in biological fluids: K. H. Lehr, P. Damm, J. Chromatogr. 526, 497 (1990). Clinical pharmacokinetics: K. Ratheiser et al., Arzneim.-Forsch. 43, 856 (1993). Toxicity study: U. Schollmeier et al., ibid. 1038. Series of articles on pharmacology and clinical efficacy: Diabetes Res. Clin. Pract. 28Suppl., S115-S149 (1995).
Properties: mp 207°.
Melting point: mp 207°
Therap-Cat: Antidiabetic.
Keywords: Antidiabetic; Sulfonylurea Derivatives.
Glimepiride
Glimepiride.svg
Clinical data
Trade names Amaryl
AHFS/Drugs.com Monograph
MedlinePlus a696016
License data
Pregnancy
category
  • US: C (Risk not ruled out)
Routes of
administration
Oral (tablets)
ATC code
Legal status
Legal status
  • US: ℞-only
  • In general: ℞ (Prescription only)
Pharmacokinetic data
Bioavailability 100%
Protein binding >99.5%
Metabolism Complete hepatic (1st stage through CYP2C9)
Biological half-life 5–8 hours
Excretion Urine (~60%), feces (~40%)
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.170.771
Chemical and physical data
Formula C24H34N4O5S
Molar mass 490.617 g/mol
3D model (JSmol)

///////////Amaryl, glimepiride, glymepiride, HOE 490

CCC1=C(CN(C1=O)C(=O)NCCC2=CC=C(C=C2)S(=O)(=O)NC(=O)NC3CCC(CC3)C)C

AMISELIMOD

$
0
0

Image result for AMISELIMOD

AMISELIMOD

UNII-358M5150LY; CAS 942399-20-4; 358M5150LY; MT-1303; Amiselimod, MT-1303

Molecular Formula: C19H30F3NO3
Molecular Weight: 377.448 g/mol

2-amino-2-[2-[4-heptoxy-3-(trifluoromethyl)phenyl]ethyl]propane-1,3-diol

Phase II Crohn’s disease; Multiple sclerosis; Plaque psoriasis

Image result for AMISELIMOD

AMISELIMOD HYDROCHLORIDE

  • Molecular FormulaC19H31ClF3NO3
  • Average mass413.902 Da
1,3-Propanediol, 2-amino-2-[2-[4-(heptyloxy)-3-(trifluoromethyl)phenyl]ethyl]-, hydrochloride (1:1)
2-Amino-2-{2-[4-(heptyloxy)-3-(trifluoromethyl)phenyl]ethyl}-1,3-propanediol hydrochloride (1:1)
942398-84-7 [RN]
MT-1303
UNII-AY898D6RU1
2-amino-2-[2-[4-(heptyloxy)-3-(trifluoromethyl)phenyl]ethyl]-1,3-propanediol, monohydrochloride
  • Originator Mitsubishi Tanabe Pharma Corporation
  • Class Propylene glycols; Small molecules
  • Mechanism of Action Immunosuppressants; Sphingosine-1-phosphate receptor antagonist

Highest Development Phases

  • Phase II Crohn’s disease; Multiple sclerosis; Plaque psoriasis
  • Phase I Autoimmune disorders; Inflammation; Systemic lupus erythematosus
  • No development reported Inflammatory bowel diseases

Most Recent Events

  • 04 Nov 2017 No recent reports of development identified for phase-I development in Autoimmune-disorders in Japan (PO, Capsule)
  • 04 Nov 2017 No recent reports of development identified for phase-I development in Autoimmune-disorders in USA (PO, Capsule)
  • 04 Nov 2017 No recent reports of development identified for phase-I development in Inflammation in Japan (PO, Capsule)
  • Image result

Amiselimod, also known as MT1303, is a potent and selective immunosuppressant and sphingosine 1 phosphate receptor modulator. Amiselimod may be potentially useful for treatment of multiple sclerosis; inflammatory diseases; autoimmune diseases; psoriasis and inflammatory bowel diseases. Amiselimod is currently being developed by Mitsubishi Tanabe Pharma Corporation

Mitsubishi Tanabe is developing amiselimod, an oral sphingosine-1-phosphate (S1P) receptor antagonist, for treating autoimmune diseases, primarily multiple sclerosis, psoriasis and inflammatory bowel diseases, including Crohn’s disease.

WO2007069712

EU states expire 2026, and

Expire in the US in June 2030 with US154 extension.

Inventors Masatoshi KiuchiKaoru MarukawaNobutaka KobayashiKunio Sugahara
Applicant Mitsubishi Tanabe Pharma Corporation

In recent years, calcineurin inhibitors such as cyclosporine FK 506 have been used to suppress rejection of patients receiving organ transplantation. While doing it, certain calcineurin inhibitors like cyclosporin can cause harmful side effects such as nephrotoxicity, hepatotoxicity, neurotoxicity, etc. For this reason, in order to suppress rejection reaction in transplant patients, development of drugs with higher safety and higher effectiveness is advanced.

[0003] Patent Documents 1 to 3 are useful as inhibitors of (acute or chronic) rejection in organ or bone marrow transplantation and also useful as therapeutic agents for various autoimmune diseases such as psoriasis and Behcet’s disease and rheumatic diseases 2 aminopropane 1, 3 dioly intermediates are disclosed.

[0004] One of these compounds, 2-amino-2- [2- (4-octylphenel) propane] 1, 3 diol hydrochloride (hereinafter sometimes referred to as FTY 720) is useful for renal transplantation It is currently under clinical development as an inhibitor of rejection reaction. FTY 720 is phosphorylated by sphingosine kinase in vivo in the form of phosphorylated FTY 720 [hereinafter sometimes referred to as FTY 720-P]. For example, 2 amino-2-phosphoryloxymethyl 4- (4-octafil-el) butanol. FTY720 – P has four types of S1 P receptors (hereinafter referred to as S1 P receptors) among five kinds of sphingosine – 1 – phosphate (hereinafter sometimes referred to as S1P) receptors It acts as an aggroove on the body (other than S1P2) (Non-Patent Document 1).

[0005] It has recently been reported that S1P1 among the S1P receptors is essential for the export of mature lymphocytes with thymus and secondary lymphoid tissue forces. FTY720 – P downregulates S1P1 on lymphocytes by acting as S1P1 ghost. As a result, the transfer of mature lymphocytes from the thymus and secondary lymphatic tissues is inhibited, and the circulating adult lymphocytes in the blood are isolated in the secondary lymphatic tissue to exert an immunosuppressive effect Has been suggested (

Non-Patent Document 2).

[0006] On the other hand, conventional 2-aminopropane 1, 3 dioly compounds are concerned as transient bradycardia expression as a side effect, and in order to solve this problem, 2-aminopropane 1, 3 diiori Many new compounds have been reported by geometrically modifying compounds. Among them, as a compound having a substituent on the benzene ring possessed by FTY 720, Patent Document 4 discloses an aminopropenol derivative as a S1P receptor modulator with a phosphate group, Patent Documents 5 and 6 are both S1P Discloses an amino-propanol derivative as a receptor modulator. However, trihaloalkyl groups such as trifluoromethyl groups are not disclosed as substituents on the benzene ring among them. In any case, it is currently the case that it has not yet reached a satisfactory level of safety as a pharmaceutical.

Patent Document 1: International Publication Pamphlet WO 94 Z 08943

Patent Document 2: International Publication Pamphlet WO 96 Z 06068

Patent Document 3: International Publication Pamphlet W 0 98 z 45 429

Patent Document 4: International Publication Pamphlet WO 02 Z 076995

Patent document 5: International public non-fret WO 2004 Z 096752

Patent Document 6: International Publication Pamphlet WO 2004 Z 110979

Non-patent document 1: Science, 2002, 296, 346-349

Non-patent document 2: Nature, 2004, 427, 355-360

Reference Example 3

5 bromo 2 heptyloxybenzonitrile

(3- 1) 5 Synthesis of bromo-2 heptyloxybenzonitrile (Reference Example Compound 3- 1)

1-Heptanol (1.55 g) was dissolved in N, N dimethylformamide (24 ml) and sodium hydride (0.321 g) was added at room temperature. After stirring for 1 hour, 5 bromo-2 fluoborosyl-tolyl (2.43 g) was added and the mixture was further stirred for 50 minutes. The reaction solution was poured into water, extracted with ethyl acetate, washed with water, saturated brine, dried over anhydrous sodium sulfate, and the solvent was distilled off under reduced pressure. After eliminating the 5 bromo 2 fluconate benzonitrile as a raw material, the reaction was carried out again under the same conditions and purification was carried out by silica gel column chromatography (hexane: ethyl acetate = 50: 1 to 5: 1) to obtain the desired product (3.10 g ) As a colorless oil.

– NMR (CDCl 3) δ (ppm): 0.89 (3H, t, J = 6.4 Hz), 1.24-1.35 (6H, m

J = 8.8 Hz), 1.48 (2H, quint, J = 7.2 Hz), 1.84 7.59 (1 H, dd, J = 8.8, 2.4 Hz), 7.65 (1 H, d, J = 2.4 Hz).

Example 1

2 Amino 2- [2- (4-heptyloxy-3 trifluoromethylph enyl) propane-1, 3-diol hydrochloride

(1 – 1) {2, 2 Dimethyl 5- [2- (4 hydroxy 3 trifluoromethylfuethyl) ethyl] 1,3 dioxane 5 mercaptothenylboronic acid t butyl ester (synthesis compound 1 1)

Reference Example Compound 2-5 (70.3 g) was dissolved in tetrahydrofuran (500 ml), t-butoxycallium (13.Og) was added, and the mixture was stirred for 1 hour. To the mixed solution was dropwise added a solution of the compound of Reference Example 1 (15.Og) in tetrahydrofuran (100 ml) under ice cooling, followed by stirring for 2 hours under ice cooling. Water was added to the reaction solution, the mixture was extracted with ethyl acetate, washed with water, saturated brine, dried with anhydrous magnesium sulfate, and the solvent was distilled off under reduced pressure. The residue was purified by silica gel column chromatography (hexane: ethyl acetate = 3: D to obtain 31. Og of a pale yellow oily matter.) The geometric isomer ratio of the obtained product was (E : Z = 1: 6).

This pale yellow oil was dissolved in ethyl acetate (200 ml), 10% palladium carbon (3.00 g) was added, and the mixture was stirred under a hydrogen atmosphere at room temperature for 7 hours. After purging the inside of the reaction vessel with nitrogen, the solution was filtered and the filtrate was concentrated. The residue was washed with diisopropyl ether to obtain the desired product (2.2 g) as a colorless powder.

1 H-NMR (CDCl 3) δ (ppm): 1. 43 (3H, s), 1.44 (3H, s), 1. 47 (9H, s), 1

(2H, m), 91- 1. 98 (2H, m), 2. 50-2.66 (2H, m), 3. 69 (2H, d, J = Il. 6 Hz), 3. 89 J = 8.2 Hz), 7. 22 (1 H, dd J = 8 Hz), 5. 02 (1 H, brs), 5. 52 . 2, 1. 7 Hz), 7. 29 (1 H, d, J = l. 7 Hz).

(1-2) {2,2 Dimethyl-5- [2- (4heptyloxy-3 trifluoromethyl) ethyl] 1,3 dioxane 5-mercaptobutyric acid t-butyl ester Synthesis (compound 1 2)

Compound 1-1 (510 mg) was dissolved in N, N dimethylformamide (10 ml), potassium carbonate (506 mg) and n-heptyl bromide (0.235 ml) were added and stirred at 80 ° C. for 2 hours. Water was added to the reaction solution, the mixture was extracted with ethyl acetate, washed with water and saturated brine, dried with anhydrous sulfuric acid

The resultant was dried with GENSCHUM and the solvent was distilled off under reduced pressure to obtain the desired product (640 mg) as a colorless oil.

– NMR (CDCl 3) δ (ppm): 0.89 (3H, t, J = 6.8 Hz), l.30-1.37 (6H, m

(2H, m), 1.91-1.98 (2H, m), 1.42-1.50 (2H, m), 1.42 (3H, s), 1.44 (3H, s), 1.47 J = 16.6 Hz), 4.00 (2H, t, J = 6.4 Hz), 4.9 8 (2H, d, J = 11.6 Hz), 3.69 1 H, brs), 6.88 (1 H, d, J = 8.5 Hz), 7.26 – 7.29 (1 H, m), 7.35 (1 H, d, J = 1.5 Hz).

(1-3) Synthesis of 2-amino-2- [2- (4heptyloxy 3 trifluoromethyl) ethyl] propane 1, 3 diol hydrochloride (Compound 1- 3)

Compound 12 (640 mg) was dissolved in ethanol (15 ml), concentrated hydrochloric acid (3 ml) was caught and stirred at 80 ° C. for 2 hours. The reaction solution was concentrated, and the residue was washed with ethyl ether to give the desired product (492 mg) as a white powder.

MS (ESI) m / z: 378 [M + H]

– NMR (DMSO-d) δ (ppm): 0.86 (3H,

6 t, J = 6.8 Hz), 1.24 – 1.39 (6

(4H, m), 3.51 (4H, d, J = 5. lHz), 4.06 (2H, m), 1.39-1.46 (2H, m), 1.68-1.78 (4H, m), 2.55-2.22 , 7.32 (2H, t, J = 5.1 Hz), 7.18 (1 H, d, J = 8.4 Hz), 7.42 – 7.45 (2 H, m), 7.76 (3 H, brs;).

PATENT

WO 2009119858

JP 2011136905

WO 2017188357

PATENT

WO-2018021517

Patent Document 1 discloses 2-amino-2- [2- (4-heptyloxy-3-trifluoromethylphenyl) ethyl] propane- 1,3 which is useful as a medicine excellent in immunosuppressive action, rejection- – diol hydrochloride is disclosed.
The production method includes the step of reducing 4-heptyloxy-3-trifluoromethylbenzoic acid (Ia) to 4-heptyloxy-3-trifluoromethylbenzyl alcohol (IIa). However, until now, there has been a problem such that the conversion is low and the by-product (IIa ‘) in which the trifluoromethyl group is reduced together with the compound (IIa) is generated in this step.
[Chemical formula 1]
 In particular, since a series of analogous substances derived from by-products (IIa ‘) are difficult to be removed in a later process, it is necessary to suppress strict production thereof in the manufacture of drug substances requiring high quality there were.

Patent Document 1: WO2007 / 069712

[Chemical formula 3]

(2-amino-2- [2- (4-heptyloxy-3-trifluoromethylphenyl) ethyl] propane- 1,3-diol hydrochloride) From
the compound (IIa), the following scheme Based on the route, 2-amino-2- [2- (4-heptyloxy-3-trifluoromethylphenyl) ethyl] propane-1,3-diol hydrochloride was prepared.
[Chemical Formula 9]

STR1
Example 2
Synthesis of 4-heptyloxy-3-trifluoromethylbenzyl chloride (Step A) A
few drops of N, N-dimethylformamide was added to a solution of compound (IIa) (26.8 g) in methylene chloride (107 mL), and 0 At 0 ° C., thionyl chloride (8.09 mL) was added dropwise. The mixture was stirred at the same temperature for 2 hours, and water (50 mL) was added to the reaction solution. The organic layer was separated and extracted, washed with water (50 mL), saturated aqueous sodium bicarbonate solution (70 mL), dried over anhydrous magnesium sulfate, and the solvent was distilled off under reduced pressure to give 4-heptyloxy-3-trifluoromethylbenzyl Chloride (28.3 g) as white crystals.
1H-NMR (CDCl 3) δ (ppm): 0.89 (3H, t, J = 6.5 Hz), 1.26-1.54 (8H, m), 1.77-1.86 (2H, m , 4.49 (2H, t, J = 6.4 Hz), 4.56 (2H, s), 6.96 (IH, d, J = 8.6 Hz), 7.49 (IH, dd, J = 2.0 Hz, 8.5 Hz), 7.58 (1 H, d, J = 1.9 Hz)
Example 3
Synthesis of dimethyl (4-heptyloxy-3-trifluoromethylbenzyl) phosphonate (Step B) To
a solution of N, N (3-trifluoromethylbenzyl ) phosphonate of 4-heptyloxy-3-trifluoromethylbenzyl chloride (6.00 g, 19.4 mmol) (2.57 g, 23.3 mmol), cesium carbonate (7.60 g, 23.3 mmol) and tetrabutylammonium iodide (7.54 g, 20.4 mmol) were added to a dimethylformamide (36 mL) And the mixture was stirred at 25 ° C. for 1 day. Toluene (36 mL) and water (18 mL) were added for phase separation, and the resulting organic layer was washed twice with a mixture of N, N-dimethylformamide (18 mL) and water (18 mL). After concentration under reduced pressure, column purification using hexane and ethyl acetate gave 4.71 g of dimethyl (4-heptyloxy-3-trifluoromethylbenzyl) phosphonate.
1
H-NMR (CDCl 3) δ (ppm): 0.89 (3 H, t, J = 6.9 Hz), 1.20 – 1.41 (6 H, m) , 1.43-1.49 (2H, m), 1.72-1.83 (2H, m), 3.09 (IH, s), 3.14 (IH, s), 3.68 (3H , 7.41 – 7.44 (2 H, t, J = 6.4 Hz), 6.94 (1 H, d, J = 8.4 Hz), 3.70 (3 H, s), 4.02 (2H, m)
Example 4
tert-Butyl (E) – {2,2-dimethyl-5- [2- (4-heptyloxy-3-trifluoromethylphenyl) vinyl] -1, 3-dioxan-5- yl} carbamate Ester synthesis (Step C) A
solution of dimethyl (1.18 g, 3.09 mmol ) (4-heptyloxy-3-trifluoromethylbenzyl) phosphonate in 1.25 mL of N, N- dimethylformamide and (2, -dimethyl-5-formyl-1,3-dioxan-5-yl) carbamic acid tert-butyl ester (961 mg, 3.71 mmol) in tetrahydrofuran (4 mL) was treated with potassium tert-butoxide (1.28 g, 4 mmol) in tetrahydrofuran (7 mL), and the mixture was stirred at 0 ° C. for 6 hours. Heptane (7 mL) and water (3 mL) were added and the layers were separated, and the obtained organic layer was washed twice with water (3 mL) and concentrated. Heptane was added and the mixture was cooled in an ice bath. The precipitated crystals were collected by filtration and dried under reduced pressure to give (E) – {2,2-dimethyl-5- [2- (4-heptyloxy- Phenyl) vinyl] -1, 3-dioxan-5-yl} carbamic acid tert-butyl ester.
1
H-NMR (CDCl 3) δ (ppm): 0.89 (3 H, t, J = 6.9 Hz), 1.29 – 1.38 (6 H, m) , 1.44 – 1.59 (17 H, m), 1.77 – 1.83 (2 H, m), 3.83 – 3.93 (2 H, m), 3.93 – 4.08 (4 H, J = 16.5 Hz), 6.48 (1 H, d, J = 16.5 Hz), 6.91 (1 H, d, J), 5.21 (1 H, brs), 6.10 J = 8.5 Hz), 7.44 (1 H, dd, J = 8.6, 2.1 Hz), 7.55 (1 H, d, J = 2.0 Hz)
Example 5
Synthesis of 2-amino-2- [2- (4-heptyloxy-3-trifluoromethylphenyl) ethyl] propane-1,3-diol hydrochloride (Step D)
(E) – {2, -dimethyl-5- [2- (4-heptyloxy-3-trifluoromethylphenyl) vinyl] -1,3-dioxan- 5-yl} carbamic acid tert-butyl ester (6.50 g, 12.6 mmol) Methanol (65 mL) solution was heated to 50 ° C., a solution of concentrated hydrochloric acid (2.55 g) in methanol (5.3 mL) was added dropwise, and the mixture was stirred at 60 ° C. for 6 hours. The mixture was cooled to around room temperature, 5% palladium carbon (0.33 g) was added thereto, and the mixture was stirred under a hydrogen gas atmosphere for 3 hours. After filtration and washing the residue with methanol (39 mL), the filtrate was concentrated and stirred at 5 ° C. for 1 hour. Water (32.5 mL) was added and the mixture was stirred at 5 ° C for 1 hour, and the precipitated crystals were collected by filtration. Washed with water (13 mL) and dried under reduced pressure to obtain 4.83 g of 2-amino-2- [2- (4-heptyloxy-3-trifluoromethylphenyl) ethyl] propane-1,3-diol hydrochloride .
MS (ESI) m / z: 378 [M + H]

Image result

PATENTS

Patent ID Patent Title Submitted Date Granted Date
US2017029378 KINASE INHIBITOR
2016-10-12
US2014296183 AMINE COMPOUND AND USE THEREOF FOR MEDICAL PURPOSES
2014-06-17
2014-10-02
Patent ID Patent Title Submitted Date Granted Date
US2017253563 KINASE INHIBITORS
2017-05-24
US9499486 Kinase inhibitor
2015-10-01
2016-11-22
US9751837 KINASE INHIBITORS
2015-10-01
2016-04-14
US8809304 Amine Compound and Use Thereof for Medical Purposes
2009-05-28
US2017209445 KINASE INHIBITORS
2015-10-01

////////////AMISELIMOD, Phase II, Crohn’s disease, Multiple sclerosis, Plaque psoriasis,  MT-1303,  MT1303,  MT 1303, Mitsubishi Tanabe Pharma Corporation, Mitsubishi , JAPAN, PHASE 2

CCCCCCCOC1=C(C=C(C=C1)CCC(CO)(CO)N)C(F)(F)F

Design, synthesis and biological evaluation of novel 5-hydroxy-2-methyl-4H-pyran-4-one derivatives as antiglioma agents

$
0
0

Med. Chem. Commun., 2018, Advance Article
DOI: 10.1039/C7MD00551B, Research Article
Yi-Bin Li, Wen Hou, Hui Lin, Ping-Hua Sun, Jing Lin, Wei-Min Chen
Two series of 5-hydroxy-2-methyl-4H-pyran-4-one derivatives were synthesized and their antiglioma activities were evaluated.

Design, synthesis and biological evaluation of novel 5-hydroxy-2-methyl-4H-pyran-4-one derivatives as antiglioma agents

Author affiliations

Abstract

D-2-Hydroxyglutarate (D-2HG) is frequently found in human brain cancers. Approximately 50–80% of grade II glioma patients have a high level of D-2HG production, which can lead to cancer initiation. In this study, a series of novel 5-hydroxy-2-methyl-4H-pyran-4-one derivatives were designed and synthesized as antiglioma agents, and their related structure–activity relationships are discussed. Among these novel compounds, 4a exhibited promising anti-proliferative activity against glioma HT1080 cells and U87 cells with an IC50 of 1.43 μM and 4.6 μM, respectively. Further studies found that the most active compound (4a) shows an 86.3% inhibitory rate against the intracellular production of D-2HG at 1 μM, and dramatic inhibitory effects, even at 1 μM on the colony formation and migration of U87 and HT1080 cells.

STR1 STR2 str3 str4
6,6′-((4-(Benzyloxy)phenyl)methylene)bis(5-hydroxy-2-methyl-4H-pyran-4- one) (4a) The reaction was performed according to the general procedure C, using 1 (1.00 g, 7.90 mmol) and 4-(benzyloxy)benzaldehyde (0.84 g, 3.95 mmol).2 The crude product was recrystallized from isopropanol affording a white powder 4a (1.53 g, 87%): mp 261.4-262.1oC; 1HNMR (300 MHz, DMSO-d6)  2.22 (s, 6H, CH3), 5.08 (s, 3H, OCH2- Ph), 5.96 (s, 1H, CH-Ar), 6.25 (s, 2H, C=CH), , 7.01 (d, J = 9.0 Hz, 2H, Ar-H3’/H5’), 7.22 (d, J = 9.0 Hz, 2H, Ar-H2’/H6’), 7.31-7.45 (m, 5H, Ph-H); 13CNMR (75 MHz, DMSO-d6)  173.95, 165.08, 158.12, 151.20, 147.68, 142.19, 140.77, 137.42, 129.87, 128.91, 128.16, 127.69, 115.46, 114.97, 111.74, 69.69, 19.63; ESI-MS m/z: 447.1 [M+H]+ ; ESI-HRMS m/z: 447.1438 [M+H]+ , calcd for C26H23O7 447.1438.

Pfizer’s Monobactam PF-?

$
0
0

STR1

Pfizer’s monobactam PF-?

1380110-34-8, C20 H24 N8 O12 S2, 632.58

Propanoic acid, 2-​[[(Z)​-​[1-​(2-​amino-​4-​thiazolyl)​-​2-​[[(2R,​3S)​-​2-​[[[[[(1,​4-​dihydro-​1,​5-​dihydroxy-​4-​oxo-​2-​pyridinyl)​methyl]​amino]​carbonyl]​amino]​methyl]​-​4-​oxo-​1-​sulfo-​3-​azetidinyl]​amino]​-​2-​oxoethylidene]​amino]​oxy]​-​2-​methyl-

2-((Z)-1-(2-Aminothiazol-4-yl)-2-((2R,3S)-2-((((1,5-dihydroxy-4-oxo-1,4-dihydropyridin-2-yl)methoxy)carbonylamino)methyl)-4-oxo-1-sulfoazetidin-3-ylamino)-2-oxoethylideneaminooxy)-2-methylpropanoic Acid

2-[[(Z)-[1-(2-Amino-4-thiazolyl)-2-[[(2R,3S)-2-[[[[[(1,4-dihydro-1,5-dihydroxy-4-oxo-2-pyridinyl)methyl]amino]carbonyl]amino]methyl]-4-oxo-1-sulfo-3-azetidinyl]amino]-2-oxoethylidene]amino]oxy]-2-methylpropanoic acid

Monobactams are a class of antibacterial agents which contain a monocyclic beta-lactam ring as opposed to a beta-lactam fused to an additional ring which is found in other beta-lactam classes, such as cephalosporins, carbapenems and penicillins. The drug Aztreonam is an example of a marketed monobactam; Carumonam is another example. The early studies in this area were conducted by workers at the Squibb Institute for Medical Research, Cimarusti, C. M. & R.B. Sykes: Monocyclic β-lactam antibiotics. Med. Res. Rev. 1984, 4, 1 -24. Despite the fact that selected

monobacatams were discovered over 25 years ago, there remains a continuing need for new antibiotics to counter the growing number of resistant organisms.

Although not limiting to the present invention, it is believed that monobactams of the present invention exploit the iron uptake mechanism in bacteria through the use of siderophore-monobactam conjugates. For background information, see: M. J. Miller, et al. BioMetals (2009), 22(1 ), 61-75.

The mechanism of action of beta-lactam antibiotics, including monobactams, is generally known to those skilled in the art and involves inhibition of one or more penicillin binding proteins (PBPs), although the present invention is not bound or limited by any theory. PBPs are involved in the synthesis of peptidoglycan, which is a major component of bacterial cell walls.

WO 2012073138

https://www.google.com/patents/WO2012073138A1?cl=en

Inventors Matthew Frank BrownSeungil HanManjinder LallMark. J. Mitton-FryMark Stephen PlummerHud Lawrence RisleyVeerabahu ShanmugasundaramJeremy T. Starr
Applicant Pfizer Inc.

Example 4, Route 1

2-({[(1Z)-1 -(2-amino-1 ,3-thiazol-4-yl)-2-({(2f?,3S)-2-[({[(1 ,5-dihydroxy-4-oxo-1 ,4- dihydropyridin-2-yl)methyl]carbamoyl}amino)methyl]-4-oxo-1 -sulfoazetidin-3- yl}amino)-2-oxoethylidene]amino}oxy)-2-methylpropanoic acid, bis sodium salt

(C92-Bis Na Salt).

Figure imgf000080_0001

C92-bis Na salt

Step 1 : Preparation of C90. A solution of C26 (16.2 g, 43.0 mmol) in tetrahydrofuran (900 mL) was treated with 1 , 1 ‘-carbonyldiimidazole (8.0 g, 47.7 mmol). After 5 minutes, the reaction mixture was treated with a solution of C9 (15 g, 25.0 mmol) in anhydrous tetrahydrofuran (600 mL) at room temperature. After 15 hours, the solvent was removed and the residue was treated with ethyl acetate (500 mL) and water (500 mL). The layers were separated and the aqueous layer was back extracted with additional ethyl acetate (300 mL). The organic layers were combined, washed with brine solution (500 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified via chromatography on silica gel (ethyl acetate / 2-propanol) to yield C90 as a yellow foam. Yield: 17.44 g, 19.62 mmol, 78%. LCMS m/z 889.5 (M+1 ). 1H NMR (400 MHz, DMSO-d6) 1 1 .90 (br s, 1 H), 9.25 (d, J=8.7 Hz, 1 H), 8.40 (br s, 1 H), 7.98 (s, 1 H), 7.50-7.54 (m, 2H), 7.32-7.47 (m, 8H), 7.28 (s, 1 H), 6.65 (br s, 1 H), 6.28 (br s, 1 H), 5.97 (s, 1 H), 5.25 (s, 2H), 5.18 (dd, J=8.8, 5 Hz, 1 H), 4.99 (s, 2H), 4.16-4.28 (m, 2H), 3.74-3.80 (m, 1 H), 3.29-3.41 (m, 1 H), 3.13-3.23 (m, 1 H), 1.42 (s, 9H), 1.41 (s, 3H), 1.39 (br s, 12H).

Step 2: Preparation of C91. A solution of C90 (8.5 g, 9.6 mmol) in anhydrous N,N- dimethylformamide (100 mL) was treated sulfur trioxide /V,/V-dimethylformamide complex (15.0 g, 98.0 mmol). The reaction was allowed to stir at room temperature for 20 minutes then quenched with water (300 mL). The resulting solid was collected by filtration and dried to yield C91 as a white solid. Yield: 8.1 g, 8.3 mmol, 87%. LCMS m/z 967.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 1 1.62 (br s, 1 H), 9.29 (d, J=8.8 Hz, 1 H), 9.02 (s, 1 H), 7.58-7.61 (m, 2H), 7.38-7.53 (m, 9H), 7.27 (s, 1 H), 7.07 (s, 1 H), 6.40 (br d, J=8 Hz, 1 H), 5.55 (s, 2H), 5.25 (s, 2H), 5.20 (dd, J=8.8, 5.6 Hz, 1 H), 4.46 (br dd, half of ABX pattern, J=17, 5 Hz, 1 H), 4.38 (br dd, half of ABX pattern, J=17, 6 Hz, 1 H), 3.92-3.98 (m, 1 H), 3.79-3.87 (m, 1 H), 3.07-3.17 (m, 1 H), 1.40 (s, 9H), 1 .39 (s, 3H), 1 .38 (s, 12H).

Step 3: Preparation of C92. A solution of C91 (8.1 g, 8.3 mmol) in anhydrous dichloromethane (200 mL) was treated with 1 M boron trichloride in p-xylenes (58.4 mL, 58.4 mmol) and allowed to stir at room temperature for 15 minutes. The reaction mixture was cooled in an ice bath, quenched with 2,2,2-trifluoroethanol (61 mL), and the solvent was removed in vacuo. A portion of the crude product (1 g) was purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.1 % formic acid modifier) to yield C92 as a white solid. Yield: 486 mg, 0.77 mmol. LCMS m/z 633.3 (M+1 ). 1H NMR (400 MHz, DMSO-d6) δ 9.22 (d, J=8.7 Hz, 1 H), 8.15 (s, 1 H), 7.26-7.42 (br s, 2H), 7.18-7.25 (m, 1 H), 6.99 (s, 1 H), 6.74 (s, 1 H), 6.32-6.37 (m, 1 H), 5.18 (dd, J=8.7, 5.7 Hz, 1 H), 4.33 (br d, J=4.6 Hz, 2H), 3.94-4.00 (m, 1 H), 3.60-3.68 (m, 1 H), 3.19-3.27 (m, 1 H), 1.40 (s, 3H), 1.39 (s, 3H).

Step 4: Preparation of C92-Bis Na Salt. A flask was charged with C92 (388 mg, 0.61 mmol) and water (5.0 mL). The mixture was cooled in an ice bath and treated dropwise with a solution of sodium bicarbonate (103 mg, 1.52 mmol) in water (5.0 mL). The sample was lyophilized to yield C92-Bis Na Salt as a white solid. Yield: 415 mg, 0.61 mmol, quantitative. LCMS m/z 633.5 (M+1 ). 1H NMR (400 MHz, D20) δ 7.80 (s, 1 H), 6.93 (s, 1 H), 6.76 (s, 1 H), 5.33 (d, J=5.7 Hz, 1 H), 4.44 (ddd, J=6.0, 6.0, 5.7 Hz, 1 H), 4.34 (AB quartet, JAB=17.7 Hz, ΔνΑΒ=10.9 Hz, 2H), 3.69 (dd, half of ABX pattern, J=14.7, 5.8 Hz, 1 H), 3.58 (dd, half of ABX pattern, J=14.7, 6.2 Hz, 1 H), 1.44 (s, 3H), 1.43 (s, 3H).

Alternate preparation of C92

Figure imgf000082_0001

Step 1 : Preparation of C93. An Atlantis pressure reactor was charged with 10% palladium hydroxide on carbon (0.375 g, John Matthey catalyst type A402028-10), C91 (0.75 g, 0.77 mmol) and treated with ethanol (35 mL). The reactor was flushed with nitrogen and pressurized with hydrogen (20 psi) for 20 hours at 20 °C. The reaction mixture was filtered under vacuum and the filtrate was concentrated using the rotary evaporator to yield C93 as a tan solid. Yield: 0.49 g, 0.62 mmol, 80%. LCMS m/z 787.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 1 1.57 (br s, 1 H), 9.27 (d, J=8.5 Hz, 1 H), 8.16 (s, 1 H), 7.36 (br s, 1 H), 7.26 (s, 1 H), 7.00 (s, 1 H), 6.40 (br s, 1 H), 5.18 (m, 1 H), 4.35 (m, 2H), 3.83 (m, 1 H), 3.41 (m, 1 H), 3.10 (m, 1 H), 1.41 (s, 6H), 1.36 (s, 18H).

Step 2: Preparation of C92. A solution of C93 (6.0 g, 7.6 mmol) in anhydrous dichloromethane (45 mL) at 0 °C was treated with trifluoroacetic acid (35.0 mL, 456 mmol). The mixture was warmed to room temperature and stirred for 2 hours. The reaction mixture was cannulated into a solution of methyl ferf-butyl ether (100 mL) and heptane (200 mL). The solid was collected by filtration and washed with a mixture of methyl ferf-butyl ether (100 mL) and heptane (200 mL) then dried under vacuum. The crude product (~5 g) was purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.1 % formic acid modifier) and lyophilized to yield C92 as a pink solid. Yield: 1.45 g, 2.29 mmol. LCMS m/z 631.0 (M-1). 1H NMR (400 MHz, DMSO-de) δ 9.20 (d, J=8.7 Hz, 1H), 8.13 (s, 1H), 7.24-7.40 (br s, 2H), 7.16-7.23 (m, 1H), 6.97 (s, 1H), 6.71 (s, 1H), 6.31-6.35 (m, 1H), 5.15 (dd, J=8.7, 5.7 Hz, 1H), 4.31 (br d, J=4.6 Hz, 2H), 3.92-3.98 (m, 1H), 3.58-3.67 (m, 1H), 3.17-3.25 (m, 1H), 1.37 (s, 3H), 1.36 (s, 3H).

Example 4, route 2

2-({[(1Z)-1-(2-amino-1,3-thiazol-4-yl)-2-({(2 ?,3S)-2-[({[(1,5-dihydroxy-4-oxo-^ dihydropyridin-2-yl)methyl]carbamoyl}amino)methyl]-4-oxo-1-sulfoazetidin-3- yl}amino)-2-oxoethylidene]amino}oxy)-2-methylpropanoic acid (C92).

lt

Figure imgf000083_0001

single

enantiomer

Figure imgf000083_0002

Step 1. Preparation of C95. A solution of C94 (50.0 g, 189.9 mmol) in

dichloromethane (100 mL) was treated with trifluoroacetic acid (50.0 mL, 661.3 mmol). The reaction mixture was stirred at room temperature for 24 hours. The dichloromethane and trifluoroacetic acid was displaced with toluene (4 x 150 mL) using vacuum, to a final volume of 120 mL. The solution was added to heptane (250 mL) and the solid was collected by filtration. The solid was washed with a mixture of toluene and heptane (1 : 3, 60 mL), followed by heptane (2 x 80 mL) and dried under vacuum at 50 °C for 19 hours to afford C95 as a solid. Yield: 30.0 g, 158 mmol, 84%. 1H NMR (400 MHz, CDCI3) δ 9.66 (s, 1 H), 7.86 – 7.93 (m, 2H), 7.73 – 7.80 (m, 2H), 4.57 (s, 2H). HPLC retention time 5.1 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μηη); column temperature 45 °C; flow rate 1.0 mL / minute; detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1.5 minutes solvent B (100%), 1.5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes.

Step 2: Preparation of C96-racemic. A solution of C95 (32.75 g; 173.1 mmol) in dichloromethane (550 mL) under nitrogen was cooled to 2 °C. The solution was treated with 2,4-dimethoxybenzylamine (28.94 g, 173.1 mmol) added dropwise over 25 minutes, maintaining the temperature below 10 °C. The solution was stirred for 10 minutes at 2 °C and then treated with molecular sieves (58.36 g, UOP Type 3A). The cold bath was removed and the reaction slurry was stirred for 3 hours at room temperature. The slurry was filtered through a pad of Celite (34.5 g) and the filter cake was rinsed with dichloromethane (135 mL). The dichloromethane filtrate (imine solution) was used directly in the following procedure.

A solution of A/-(ferf-butoxycarbonyl)glycine (60.6 g, 346.1 mmol) in

tetrahydrofuran (622 mL) under nitrogen was cooled to -45 °C and treated with triethylamine (38.5 g, 380.8 mmol). The mixture was stirred for 15 minutes at -45 °C and then treated with ethyl chloroformate (48.8 g, 450 mmol) over 15 minutes. The reaction mixture was stirred at -50 °C for 7 hours. The previously prepared imine solution was added via an addition funnel over 25 minutes while maintaining the reaction mixture temperature below -40 °C. The slurry was treated with triethylamine (17.5 g, 173 mmol) and the reaction mixture was slowly warmed to room temperature over 5 hours and stirred for an additional 12 hours. The reaction slurry was charged with water (150 mL) and the volatiles removed using a rotary evaporator. The reaction mixture was charged with additional water (393 mL) and the volatiles removed using a rotary evaporator. The mixture was treated with methyl ferf-butyl ether (393 mL) and vigorously stirred for 1 hour. The solid was collected by vacuum filtration and the filter cake was rinsed with a mixture of methyl ferf-butyl ether and water (1 : 1 , 400 mL). The solid was collected and dried in a vacuum oven at 50 °C for 16 hours to afford C96- racemic. Yield: 55.8 g, 1 13 mmol, 65%. 1H-NMR (400 MHz, DMSO-d6) δ 7.85 (s, NH), 7.80 (s, 4H), 6.78 (d, J=7.8 Hz, 1 H), 6.25 (m, 1 H), 6.10 (m, 1 H), 4.83 (m, 1 H), 4.38 (d, J=9.5 Hz, 1 H), 3.77-3.95 (m, 3H), 3.62 (s, 3H), 3.45 (m, 1 H), 3.40 (s, 3H), 1.38 (s, 9H). HPLC retention time 6.05 minutes; XBridge C8 column (4.6 x 75 mm, 3.5 μηη); column temperature 45 °C; flow rate 2.0 mL/minute; detection UV 210 nm, 230 nm, and 254 nm; mobile phase: solvent A = methanesulfonic acid (5%) in 10 mmol sodium octylsulfonate, solvent B = acetonitrile (100%); gradient elusion: 0-1.5 minutes solvent A (95%) and solvent B (5%), 1.5-8.5 minutes solvent A (5%) and solvent B (95%), 8.5- 10.0 minutes solvent A (5%) and solvent B (95%), 10.01 -12.0 minutes solvent A (95%) and solvent B (5%); total run time 12.0 minutes.

Step 3: Preparation of C97-racemic. A solution of C96-racemic (15.0 g, 30.3 mmol) in ethyl acetate (150 mL) under nitrogen was treated with ethanolamine (27.3 mL, 454.1 mmol). The reaction mixture was heated at 90 °C for 3 hours and then cooled to room temperature. The mixture was charged with water (150 mL) and the layers separated. The aqueous layer was extracted with ethyl acetate (75 mL) and the combined organic layers washed with water (2 x 150 mL) followed by saturated aqueous sodium chloride (75 mL). The organic layer was dried over magnesium sulfate, filtered and the filtrate concentrated to a volume of 38 mL. The filtrate was treated with heptane (152 mL) and the solid was collected by filtration. The solid was washed with heptane and dried at 50 °C in a vacuum oven overnight to yield C97-racemic as a solid. Yield: 9.68 g, 26.5 mmol, 88%. LCMS m/z 967.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 7.64 (d, J=9.4 Hz, 1 H), 7.14 (d, J=8.2 Hz, 1 H), 6.56 (s, 1 H), 6.49 (dd, J=8.20, 2.3 Hz, 1 H), 4.78 (dd, J=9.37, 5.1 Hz, 1 H), 4.30 (d, J=14.8 Hz, 1 H), 4.14 (d, J=14.8 Hz, 1 H), 3.77 (s, 3H), 3.75 (s, 3H), 3.45 – 3.53 (m, 1 H), 2.65 – 2.75 (m, 1 H), 2.56 – 2.64 (m, 1 H), 1.38 (s, 9H), 1.30 – 1.35 (m, 2H). HPLC retention time 5.1 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μΐη); column temperature 45 °C; flow rate 1.0 mL / minute;

detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1 .5 minutes solvent B (100%), 1 .5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes. Step 4: Preparation of C97-(2R,3S) enantiomer. A solution of C97-racemic (20.0 g, 54.7 mmol) in ethyl acetate (450 mL) was treated with diatomaceous earth (5.0 g) and filtered through a funnel charged with diatomaceous earth. The filter cake was washed with ethyl acetate (150 mL). The filtrate was charged with diatomaceous earth (20.0 g) and treated with (-)-L-dibenzoyltartaric acid (19.6 g, 54.7 mmol). The slurry was heated at 60 °C for 1.5 hours and then cooled to room temperature. The slurry was filtered and the solid washed with ethyl acetate (90 mL). The solid was collected and dried at 50 °C in a vacuum oven for 17 hours to yield C97-(2R,3S) enantiomer as a solid (mixed with diatomaceous earth). Yield: 17.3 g, 23.9 mmol, 43.6%, 97.6% ee. 1H NMR (400 MHz, DMSO-de) δ 7.89 – 7.91 (m, 4H), 7.59 – 7.65 (m, 3H), 7.44 – 7.49 (m, 4H), 7.09 (d, J=8.3 Hz, 1 H), 6.53 (d, J=2.3 Hz, 1 H), 6.49 (dd, J=8.3, 2.3 Hz, 1 H), 5.65 (s, 2H), 4.85 (dd, J=9.3, 4.9 Hz, 1 H), 4.30 (d, J=15.3 Hz, 1 H), 4.10 (d, J=15.3 Hz, 1 H), 3.74 (s, 3H), 3.72 (s, 3H), 3.68 – 3.70 (m, 1 H), 2.92 – 2.96 (dd, J=13.6, 5.4 Hz, 1 H), 2.85 – 2.90 (dd, J=13.6, 6.3 Hz, 1 H), 1.36 (s, 9H). HPLC retention time 5.1 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μηη); column temperature 45 °C; flow rate 1.0 mL / minute; detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1 .5 minutes solvent B (100%), 1.5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes. Chiral HPLC retention time 9.1 minutes; column: Chiralcel OD-H column (250 mm x 4.6 mm); column temperature 40 °C; flow rate 1 .0 mL / minute; detection UV 208 nm; mobile phase: solvent A = ethanol (18%), solvent B = heptane (85%); isocratic elusion; total run time 20.0 minutes.

Step 5: Preparation of C98-(2R,3S) enantiomer. A solution of C97-(2R,3S) enantiomer. (16.7 g, 23.1 mmol) in ethyl acetate (301 mL) was treated with diatomaceous earth (18.3 g) and 5% aqueous potassium phosphate tribasic (182 mL). The slurry was stirred for 30 minutes at room temperature, then filtered under vacuum and the filter cake washed with ethyl acetate (2 x 67 mL). The filtrate was washed with 5% aqueous potassium phosphate tribasic (18 mL) and the organic layer dried over magnesium sulfate. The solid was filtered and the filter cake washed with ethyl acetate (33 mL). The filtrate was concentrated to a volume of 42 mL and slowly added to heptane (251 mL) and the resulting solid was collected by filtration. The solid was washed with heptane and dried at 50 °C in a vacuum oven for 19 hours to yield C98- (2R,3S) enantiomer as a solid. Yield: 6.4 g, 17.5 mmol, 76%, 98.8% ee. 1H NMR (400 MHz, DMSO-de) δ 7.64 (d, J=9.4 Hz, 1 H), 7.14 (d, J=8.2 Hz, 1 H), 6.56 (s, 1 H), 6.49 (dd, J=8.20, 2.3 Hz, 1 H), 4.78 (dd, J=9.37, 5.1 Hz, 1 H), 4.30 (d, J=14.8 Hz, 1 H), 4.14 (d, J=14.8 Hz, 1 H), 3.77 (s, 3H), 3.75 (s, 3H), 3.45 – 3.53 (m, 1 H), 2.65 – 2.75 (m, 1 H), 2.56 – 2.64 (m, 1 H), 1.38 (s, 9H), 1.30 – 1.35 (m, 2H). HPLC retention time 5.2 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μηη); column temperature 45 °C; flow rate 1.0 mL / minute; detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1 .5 minutes solvent B (100%), 1.5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes. Chiral HPLC retention time 8.7 minutes; column: Chiralcel OD-H column (250 mm x 4.6 mm); column temperature 40 °C; flow rate 1.0 mL / minute; detection UV 208 nm; mobile phase: solvent A = ethanol (18%), solvent B = heptane (85%); isocratic elusion; total run time 20.0 minutes.

Step 6: Preparation of C99. A solution of potassium phosphate tribasic N-hydrate (8.71 g, 41 .05 mmol) in water (32.0 mL) at 22 °C was treated with a slurry of C26- mesylate salt (12.1 g, 27.4 mmol, q-NMR potency 98%) in dichloromethane (100.00 mL). The slurry was stirred for 1 hour at 22 °C. The reaction mixture was transferred to a separatory funnel and the layers separated. The aqueous layer was back extracted with dichloromethane (50.0 mL). The organic layers were combined, dried over magnesium sulfate, filtered under vacuum and the filter cake washed with

dichloromethane (2 x 16 mL). The filtrate (-190 mL, amine solution) was used directly in the next step.

A solution of 1 ,1 ‘-carbonyldiimidazole (6.66 g, 41 .0 mmol) in dichloromethane (100 mL) at 22 °C under nitrogen was treated with the previously prepared amine solution (-190 mL) added dropwise using an addition funnel over 3 hour at 22 °C with stirring. After the addition, the mixture was stirred for 1 hour at 22 °C, then treated with C98-(2R,3S) enantiomer. (10.0 g, 27.4 mmol) followed by /V,/V-dimethylformamide (23.00 mL). The reaction mixture was stirred at 22 °C for 3 hours and then heated at 40 °C for 12 hours. The solution was cooled to room temperature and the dichloromethane was removed using the rotary evaporator. The reaction mixture was diluted with ethyl acetate (216.0 mL) and washed with 10% aqueous citric acid (216.0 mL), 5% aqueous sodium chloride (2 x 216.0 mL), dried over magnesium sulfate and filtered under vacuum. The filter cake was washed with ethyl acetate (3 x 13 mL) and the ethyl acetate solution was concentrated on the rotary evaporator to a volume of (-1 10.00 mL) providing a suspension. The suspension (~1 10.00 mL) was warmed to 40 °C and transferred into a stirred solution of heptane (22 °C) over 1 hour, to give a slurry. The slurry was stirred for 1 hour and filtered under vacuum. The filter cake was washed with heptane (3 x 30 mL) and dried under vacuum at 50 °C for 12 hours to afford C99 as a solid. Yield: 18.1 g, 24.9 mmol, 92%. LCMS m/z 728.4 (M+1 ). 1H NMR (400 MHz, DMSO-d6) δ 8.09 (s, 1 H), 7.62 (d, J=9.4 Hz, 1 H), 7.33-7.52 (m, 10H), 7.07 (d, J=8.3 Hz, 1 H), 6.51 (d, J=2.3 Hz, 1 H), 6.50 (m, 1 H), 6.44 (dd, J=8.3, 2.3 Hz, 1 H), 6.12 (m, 1 H), 6.07 (s, 1 H), 5.27 (s, 2H), 5.00 (s, 2H), 4.73 (dd, J=9.4, 5.2 Hz, 1 H), 4.38 (d, J=15.0 Hz, 1 H), 4.19 (m, 2H), 3.99 (d, J=15.0 Hz, 1 H), 3.72 (s, 3H), 3.71 (s, 3H), 3.48 (m, 1 H), 3.28 (m, 1 H), 3.12 (m, 1 H), 1 .37 (s, 9H).

Step 7: Preparation of C100. A solution of C99 (46.5 g, 63.9 mmol) in acetonitrile (697 mL and water (372 mL) was treated with potassium persulfate (69.1 g, 255.6 mmol) and potassium phosphate dibasic (50.1 g, 287.5 mmol). The biphasic mixture was heated to 75 °C and vigorously stirred for 1.5 hours. The pH was maintained between 6.0-6.5 by potassium phosphate dibasic addition (-12 g). The mixture was cooled to 20 °C, the suspension was filtered and washed with acetonitrile (50 mL). The filtrate was concentrated using the rotary evaporator and treated with water (50 mL) followed by ethyl acetate (200 mL). The slurry was stirred for 2 hours at room temperature, filtered and the solid dried under vacuum at 40 °C overnight. The solid was slurried in a mixture of ethyl acetate and water (6 : 1 , 390.7 mL) at 20 °C for 1 hour then collected by filtration. The solid was dried in a vacuum oven to yield C100. Yield: 22.1 g, 38.3 mmol, 60%. 1H NMR (400 MHz, DMSO-d6) δ 8.17 (br s, 1 H), 7.96 (s, 1 H), 7.58 (d, J=9.6 Hz, 1 H), 7.29-7.50 (m, 10H), 6.49 (dd, J=8.0, 6.0 Hz, 1 H), 6.08 (dd, J=5.6, 5.2 Hz, 1 H), 5.93 (s, 1 H), 5.22 (s, 2H), 4.96 (s, 2H), 4.77 (dd, J=9.6, 5.0 Hz, 1 H), 4.16 (m, 2H), 3.61 (m, 1 H), 3.1 1 (m, 2H), 1.36 (s, 9H). HPLC retention time 6.17 minutes; XBridge C8 column (4.6 x 75 mm, 3.5 μηη); column temperature 45 °C; flow rate 2.0 mL/minute; detection UV 210 nm, 230 nm, and 254 nm; mobile phase: solvent A = methanesulfonic acid (5%) in 10 mmol sodium octylsulfonate, solvent B = acetonitrile (100%); gradient elusion: 0-1 .5 minutes solvent A (95%) and solvent B (5%), 1.5-8.5 minutes solvent A (5%) and solvent B (95%), 8.5-10.0 minutes solvent A (5%) and solvent B (95%), 10.01- 12.0 minutes solvent A (95%) and solvent B (5%); total run time 12.0 minutes.

Step 8: Preparation of C101. A solution of trifluoroacetic acid (120 mL, 1550 mmol) under nitrogen was treated with methoxybenzene (30 mL, 269 mmol) and cooled to -5 °C. Solid C100 (17.9 g, 31.0 mmol) was charged in one portion at -5 °C and the resulting mixture stirred for 3 hours. The reaction mixture was cannulated with nitrogen pressure over 15 minutes to a stirred mixture of Celite (40.98 g) and methyl ferf-butyl ether (550 mL) at 10 °C. The slurry was stirred at 16 °C for 30 minutes, then filtered under vacuum. The filter cake was rinsed with methyl ferf-butyl ether (2 x 100 mL). The solid was collected and slurried in methyl ferf-butyl ether (550 mL) with vigorous stirring for 25 minutes. The slurry was filtered by vacuum filtration and washed with methyl ferf-butyl ether (2 x 250 mL). The solid was collected and dried in a vacuum oven at 60 °C for 18 hours to afford C101 on Celite. Yield: 57.6 g total = C101 + Celite; 16.61 g C101 , 28.1 mmol, 91%. 1H NMR (400 MHz, DMSO-d6) δ 8.75-8.95 (br s, 2H), 8.65 (s, 1 H), 8.21 (s, 1 H), 7.30-7.58 (m, 10H), 6.83 (br s, 1 H), 6.65 (br s, 1 H), 6.17 (s, 1 H), 5.30 (s, 2H), 5.03 (s, 2H), 4.45 (br s, 1 H), 4.22 (br s, 2H), 3.77 (m, 1 H), 3.36 (m, 1 H), 3.22 (m, 1 H). 19F NMR (376 MHz, DMSO-d6) δ -76.0 (s, 3F). HPLC retention time 5.81 minutes; XBridge C8 column (4.6 x 75 mm, 3.5 μηη); column temperature 45 °C; flow rate 2.0 mL/minute; detection UV 210 nm, 230 nm, and 254 nm; mobile phase: solvent A = methanesulfonic acid (5%) in 10 mmol sodium octylsulfonate, solvent B = acetonitrile (100%); gradient elusion: 0-1.5 minutes solvent A (95%) and solvent B (5%), 1.5-8.5 minutes solvent A (5%) and solvent B (95%), 8.5-10.0 minutes solvent A (5%) and solvent B (95%), 10.01-12.0 minutes solvent A (95%) and solvent B (5%); total run time 12.0 minutes.

Step 9: Preparation of C90. A suspension of C101 (67.0 g, 30% activity on Celite = 33.9 mmol) in acetonitrile (281 .4 mL) was treated with molecular sieves 4AE (40.2 g), C5 (17.9 g, 33.9 mmol), 4-dimethylaminopyridine (10.4 g, 84.9 mmol) and the mixture was stirred at 40°C for 16 hours. The reaction mixture was cooled to 20 °C, filtered under vacuum and the filter cake washed with acetonitrile (2 x 100 mL). The filtrate was concentrated under vacuum to a volume of -50 mL. The solution was diluted with ethyl acetate (268.0 mL) and washed with 10% aqueous citric acid (3 x 134 mL) followed by 5% aqueous sodium chloride (67.0 mL). The organic layer was dried over magnesium sulfate and filtered under vacuum. The filter cake was washed with ethyl acetate (2 x 50 mL) and the filtrate was concentrated to a volume of -60 mL. The filtrate was added slowly to heptane (268 mL) with stirring and the slurry was stirred at 20 °C for 1 hour. The slurry was filtered under vacuum and the filter cake washed with a mixture of heptane and ethyl acetate (4: 1 , 2 x 27 mL). The solid was collected and dried under vacuum for 12 hours at 50 °C to afford a solid. The crude product was purified via chromatography on silica gel (ethyl acetate / 2-propanol), product bearing fractions were combined and the volume was reduced to -60 mL. The solution was added dropwise to heptane (268 mL) with stirring. The slurry was stirred at room temperature for 3 hours, filtered and washed with heptane and ethyl acetate (4: 1 , 2 x 27 mL). The solid was collected and dried under vacuum for 12 hours at 50 °C to afford C90 as a solid. Yield: 16.8 g, 18.9 mmol, 58%. LCMS m/z 889.4 (M+1 ). 1H NMR (400 MHz, DMSO-cfe) 1 1.90 (br s, 1 H), 9.25 (d, J=8.7 Hz, 1 H), 8.40 (br s, 1 H), 7.98 (s, 1 H), 7.50-7.54 (m, 2H), 7.32- 7.47 (m, 8H), 7.28 (s, 1 H), 6.65 (br s, 1 H), 6.28 (br s, 1 H), 5.97 (s, 1 H), 5.25 (s, 2H), 5.18 (dd, J=8.8, 5 Hz, 1 H), 4.99 (s, 2H), 4.16-4.28 (m, 2H), 3.74-3.80 (m, 1 H), 3.29-3.41 (m, 1 H), 3.13-3.23 (m, 1 H), 1 .42 (s, 9H), 1 .41 (s, 3H), 1.39 (br s, 12H).

Step 10: Preparation of C91. A solution of C90 (14.5 g, 16.3 mmol) in anhydrous N,N- dimethylformamide (145.0 mL) was treated with sulfur trioxide /V,/V-dimethylformamide complex (25.0 g, 163.0 mmol). The reaction mixture was stirred at room temperature for 45 minutes, then transferred to a stirred mixture of 5% aqueous sodium chloride (290 mL) and ethyl acetate (435 mL) at 0 °C. The mixture was warmed to 18 °C and the layers separated. The aqueous layer was extracted with ethyl acetate (145 mL) and the combined organic layers washed with 5% aqueous sodium chloride (3 x 290 mL) followed by saturated aqueous sodium chloride (145 mL). The organic layer was dried over magnesium sulfate, filtered through diatomaceous earth and the filter cake washed with ethyl acetate (72 mL). The filtrate was concentrated to a volume of 36 mL and treated with methyl ferf-butyl ether (290 mL), the resulting slurry was stirred at room temperature for 1 hour. The solid was collected by filtration, washed with methyl ferf- butyl ether (58 mL) and dried at 50 °C for 2 hours followed by 20 °C for 65 hours in a vacuum oven to yield C91 as a solid. Yield: 15.0 g, 15.4 mmol, 95%. LCMS m/z 967.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 1 1.62 (br s, 1 H), 9.29 (d, J=8.8 Hz, 1 H), 9.02 (s, 1 H), 7.58-7.61 (m, 2H), 7.38-7.53 (m, 9H), 7.27 (s, 1 H), 7.07 (s, 1 H), 6.40 (br d, J=8.0 Hz, 1 H), 5.55 (s, 2H), 5.25 (s, 2H), 5.20 (dd, J=8.8, 5.6 Hz, 1 H), 4.46 (br dd, half of ABX pattern, J=17.0, 5.0 Hz, 1 H), 4.38 (br dd, half of ABX pattern, J=17.0, 6.0 Hz, 1 H), 3.92- 3.98 (m, 1 H), 3.79-3.87 (m, 1 H), 3.07-3.17 (m, 1 H), 1.40 (s, 9H), 1.39 (s, 3H), 1.38 (s, 12H).

Step 11 : Preparation of C92. A solution of C91 (20.0 g, 20.6 mmol) in

dichloromethane (400 mL) was concentrated under reduced pressure (420 mmHg) at 45 °C to a volume of 200 mL. The solution was cooled to -5 °C and treated with 1 M boron trichloride in dichloromethane (206.0 mL, 206.0 mmol) added dropwise over 40 minutes. The reaction mixture was warmed to 15 °C over 1 hour with stirring. The slurry was cooled to -15 °C and treated with a mixture of 2,2,2-trifluoroethanol (69.2 mL) and methyl ferf-butyl ether (400 mL), maintaining the temperature at -15 °C. The reaction mixture was warmed to 0 °C over 1 hour. The suspension was filtered using nitrogen pressure and the solid washed with methyl ferf-butyl ether (2 x 200 mL).

Nitrogen was passed over the solid for 2 hours. The solid was collected and suspended in methyl ferf-butyl ether (400 mL) for 1 hour with stirring at 18 °C. The suspension was filtered using nitrogen pressure and the solid washed with methyl ferf-butyl ether (2 x 200 mL). Nitrogen was passed over the resulting solid for 12 hours. A portion of the crude product was neutralized with 1 M aqueous ammonium formate to pH 5.5 with minimal addition of /V,/V-dimethylformamide to prevent foaming. The feed solution was filtered and purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.2% formic acid modifier). The product bearing fractions were combined and concentrated to remove acetonitrile. The solution was captured on a GC-161 M column, washed with deionized water and blown dry with nitrogen pressure. The product was released using a mixture of methanol / water (10: 1 ) and the product bearing fractions were added to a solution of ethyl acetate (6 volumes). The solid was collected by filtration to afford C92 as a solid. Yield: 5.87 g, 9.28 mmol. LCMS m/z 633.3 (M+1 ). 1H NMR (400 MHz, DMSO-d6) δ 9.22 (d, J=8.7 Hz, 1 H), 8.15 (s, 1 H), 7.26-7.42 (br s, 2H), 7.18-7.25 (m, 1 H), 6.99 (s, 1 H), 6.74 (s, 1 H), 6.32-6.37 (m, 1 H), 5.18 (dd, J=8.7, 5.7 Hz, 1 H), 4.33 (br d, J=4.6 Hz, 2H), 3.94-4.00 (m, 1 H), 3.60-3.68 (m, 1 H), 3.19-3.27 (m, 1 H), 1.40 (s, 3H), 1.39 (s, 3H).

PAPER

Journal of Medicinal Chemistry (2014), 57(9), 3845-3855

Siderophore Receptor-Mediated Uptake of Lactivicin Analogues in Gram-Negative Bacteria

Medicinal Chemistry, Computational Chemistry, §Antibacterials Research Unit, and Structural Biology, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
J. Med. Chem.201457 (9), pp 3845–3855
DOI: 10.1021/jm500219c
Publication Date (Web): April 2, 2014
Copyright © 2014 American Chemical Society
*Phone: (860)-686-1788. E-mail: seungil.han@pfizer.com.

Abstract

Abstract Image

Multidrug-resistant Gram-negative pathogens are an emerging threat to human health, and addressing this challenge will require development of new antibacterial agents. This can be achieved through an improved molecular understanding of drug–target interactions combined with enhanced delivery of these agents to the site of action. Herein we describe the first application of siderophore receptor-mediated drug uptake of lactivicin analogues as a strategy that enables the development of novel antibacterial agents against clinically relevant Gram-negative bacteria. We report the first crystal structures of several sideromimic conjugated compounds bound to penicillin binding proteins PBP3 and PBP1a from Pseudomonas aeruginosa and characterize the reactivity of lactivicin and β-lactam core structures. Results from drug sensitivity studies with β-lactamase enzymes are presented, as well as a structure-based hypothesis to reduce susceptibility to this enzyme class. Finally, mechanistic studies demonstrating that sideromimic modification alters the drug uptake process are discussed.

PAPER

Pyridone-Conjugated Monobactam Antibiotics with Gram-Negative Activity

Worldwide Medicinal Chemistry, Computational Chemistry, §Antibacterials Research Unit, Pharmacokinetics, Dynamics & Metabolism, Structural Biology, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
J. Med. Chem.201356 (13), pp 5541–5552
DOI: 10.1021/jm400560z
Publication Date (Web): June 11, 2013
Copyright © 2013 American Chemical Society
*Phone: 860-441-3522. E-mail: matthew.f.brown@pfizer.com.
Abstract Image

Herein we describe the structure-aided design and synthesis of a series of pyridone-conjugated monobactam analogues with in vitro antibacterial activity against clinically relevant Gram-negative species including Pseudomonas aeruginosaKlebsiella pneumoniae, and Escherichia coli. Rat pharmacokinetic studies with compound 17 demonstrate low clearance and low plasma protein binding. In addition, evidence is provided for a number of analogues suggesting that the siderophore receptors PiuA and PirA play a role in drug uptake in P. aeruginosa strain PAO1.

STR1

17 as a solid. Yield: 5.87 g, 9.28 mmol. LCMS m/z 633.3 (M+1). 1H NMR (400 MHz, DMSOd6) δ 9.22 (d, J=8.7 Hz, 1H), 8.15 (s, 1H), 7.26-7.42 (br s, 2H), 7.18-7.25 (m, 1H), 6.99 (s, 1H), 6.74 (s, 1H), 6.32-6.37 (m, 1H), 5.18 (dd, J=8.7, 5.7 Hz, 1H), 4.33 (br d, J=4.6 Hz, 2H), 3.94-4.00 (m, 1H), 3.60-3.68 (m, 1H), 3.19-3.27 (m, 1H), 1.40 (s, 3H), 1.39 (s, 3H).

Nc1nc(cs1)\C(=N\OC(C)(C)C(=O)O)C(=O)N[C@@H]3C(=O)N([C@@H]3CNC(=O)NCC2=CC(=O)C(O)=CN2O)S(=O)(=O)O

PAPER

Process Development for the Synthesis of Monocyclic β-Lactam Core 17

Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00359
Publication Date (Web): January 4, 2018
Copyright © 2018 American Chemical Society
Abstract Image

Process development and multikilogram synthesis of the monocyclic β-lactam core 17 for a novel pyridone-conjugated monobactam antibiotic is described. Starting with commercially available 2-(2,2-diethoxyethyl)isoindoline-1,3-dione, the five-step synthesis features several telescoped operations and direct isolations to provide significant improvement in throughput and reduced solvent usage over initial scale-up campaigns. A particular highlight in this effort includes the development of an efficient Staudinger ketene–imine [2 + 2] cycloaddition reaction of N-Boc-glycine ketene 12 and imine 9 to form racemic β-lactam 13 in good isolated yield (66%) and purity (97%). Another key feature in the synthesis involves a classical resolution of racemic amine 15 to afford single enantiomer salt 17 in excellent isolated yield (45%) with high enantiomeric excess (98%).

Figure

https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.7b00359/suppl_file/op7b00359_si_001.pdf

Nc1nc(cs1)\C(=N\OC(C)(C)C(=O)O)C(=O)N[C@@H]3C(=O)N([C@@H]3CNC(=O)NCC2=CC(=O)C(O)=CN2O)S(=O)(=O)O

////////////////////////////////////////////////////////////////////////

J. Med. Chem.201356 (13), pp 5541–5552
DOI: 10.1021/jm400560z

OXYGEN ANALOGUE…………..

STR2
 1380110-45-1, C20 H23 N7 O13 S2, 633.57
Propanoic acid, 2-​[[(Z)​-​[1-​(2-​amino-​4-​thiazolyl)​-​2-​[[(2R,​3S)​-​2-​[[[[(1,​4-​dihydro-​1,​5-​dihydroxy-​4-​oxo-​2-​pyridinyl)​methoxy]​carbonyl]​amino]​methyl]​-​4-​oxo-​1-​sulfo-​3-​azetidinyl]​amino]​-​2-​oxoethylidene]​amino]​oxy]​-​2-​methyl-
2-[[(Z)-[1-(2-Amino-4-thiazolyl)-2-[[(2R,3S)-2-[[[[(1,4-dihydro-1,5-dihydroxy-4-oxo-2-pyridinyl)methoxy]carbonyl]amino]methyl]-4-oxo-1-sulfo-3-azetidinyl]amino]-2-oxoethylidene]amino]oxy]-2-methylpropanoic acid

STR2

18 as a light yellow solid. Yield: 43 mg, 0.068 mmol, 51%. LCMS m/z 634.4 (M+1). 1H NMR (400 MHz, DMSO-d6), characteristic peaks: δ 9.29 (d, J=8.5 Hz, 1H), 8.10 (s, 1H), 7.04-7.10 (m, 1H), 7.00 (s, 1H), 6.75 (s, 1H), 5.05-5.30 (m, 3H), 4.00-4.07 (m, 1H), 1.42 (s, 3H), 1.41 (s, 3H).

Nc1nc(cs1)\C(=N\OC(C)(C)C(=O)O)C(=O)N[C@@H]3C(=O)N([C@@H]3CNC(=O)OCC2=CC(=O)C(O)=CN2O)S(=O)(=O)O

Step 4: Preparation of 18-Bis Na salt. A suspension of 5 (212 mg, 0.33 mmol) in water (10 mL) was cooled to 0 oC and treated with a solution of sodium bicarbonate (56.4 mg, 0.67 mmol) in water (2 mL), added dropwise. The reaction mixture was cooled to -70 oC (frozen) and lyophilized to afford 18-Bis Na salt as a white solid. Yield: 210 mg, 0.31 mmol, 93%. LCMS m/z 632.5 (M-1). 1H NMR (400 MHz, D2O) δ 7.87 (s, 1H), 6.94 (s, 1H), 6.92 (s, 1H), 5.35 (d, J=5 Hz, 1H), 5.16 (s, 2H), 4.46-4.52 (m, 1H), 3.71 (dd, half of ABX pattern, J=14.5, 6 Hz, 1H), 3.55 (dd, half of ABX pattern, J=14.5, 6 Hz, 1H), 1.43 (s, 3H), 1.42 (s, 3H).

WO 2012073138

Inventors Matthew Frank BrownSeungil HanManjinder LallMark. J. Mitton-FryMark Stephen PlummerHud Lawrence RisleyVeerabahu ShanmugasundaramJeremy T. Starr
Applicant Pfizer Inc.

Example 5

disodium 2-({[(1Z)-1 -(2-amino-1 ,3-thiazol-4-yl)-2-({(2R,3S)-2-[({[(1 ,5-dihydroxy-4- oxo-1 ,4-dihydropyridin-2-yl)methoxy]carbonyl}amino)methyl]-4-oxo-1 – sulfonatoazetidin-3-yl}amino)-2-oxoethylidene]amino}oxy)-2-methylpropanoate

(C104-Bis Na salt).

Figure imgf000092_0001

Step 1 : Preparation of C102. A solution of C28 (300 mg, 0.755 mmol) in

tetrahydrofuran (10 mL) was treated with 1 , 1 ‘-carbonyldiimidazole (379 mg, 2.26 mmol) at room temperature and stirred for 20 hours. The yellow reaction mixture was treated with a solution of C9 (286 mg, 0.543 mmol) in tetrahydrofuran (25 mL). The mixture was stirred for 6 hours at room temperature, then treated with water (20 mL) and extracted with ethyl acetate (3 x 25 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified via chromatography on silica gel (heptane / ethyl acetate / 2-propanol) to afford C102 as a light yellow solid. Yield: 362 mg, 0.381 mmol, 62%. LCMS m/z 950.4 (M+1 ). 1H NMR (400 MHz, DMSO-de), characteristic peaks: δ 9.31 (d, J=8.4 Hz, 1 H), 8.38 (s, 1 H), 8.00 (s, 1 H), 7.41 (br d, J=8.2 Hz, 2H), 7.36 (br d, J=8.8 Hz, 2H), 7.26 (s, 1 H), 6.10 (s, 1 H), 5.20 (s, 2H), 4.92 (br s, 4H), 3.77 (s, 3H), 3.76 (s, 3H), 1.45 (s, 9H), 1.38 (s, 9H). Step 2: Preparation of C103. A solution of C102 (181 mg, 0.191 mmol) in anhydrous /V,/V-dimethylformamide (2.0 mL) was treated with sulfur trioxide pyridine complex (302 mg, 1.91 mmol). The reaction mixture was allowed to stir at room temperature for 6 hours, then cooled to 0 °C and quenched with water. The resulting solid was collected by filtration and dried in vacuo to yield C103 as a white solid. Yield: 145 mg, 0.14 mmol, 74%. APCI m/z 1028.5 (M-1 ). 1H NMR (400 MHz, DMSO-d6), characteristic peaks: δ 1 1.65 (br s, 1 H), 9.37 (d, J=8.6 Hz, 1 H), 8.87 (s, 1 H), 7.49 (br d, J=8.6 Hz, 2H), 7.43 (br d, J=8.6 Hz, 2H), 7.26 (s, 1 H), 7.01 (br d, J=8.9 Hz, 2H), 7.00 (br d, J=8.8 Hz, 2H), 5.43 (s, 2H), 5.20 (dd, J=8.4, 6 Hz, 1 H), 4.01-4.07 (m, 1 H), 3.78 (s, 3H), 3.77 (s, 3H), 3.50- 3.58 (m, 1 H), 3.29-3.37 (m, 1 H), 1.44 (s, 9H), 1.37 (s, 9H). Step 3: Preparation of C104. A solution of C103 (136 mg, 0.132 mmol) in anhydrous dichloromethane (5 mL) was treated with 1 M boron trichloride in p-xylenes (0.92 mL, 0.92 mmol) and allowed to stir at room temperature for 40 minutes. The reaction mixture was cooled in an ice bath, quenched with water (0.4 mL), and transferred into a solution of methyl ferf-butyl ether: heptane (1 :2, 12 mL). The solvent was removed in vacuo and the crude product was purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.1 % formic acid modifier) to yield C104 as a light yellow solid. Yield: 43 mg, 0.068 mmol, 51 %. LCMS m/z 634.4 (M+1 ). 1H NMR (400 MHz, DMSO-de), characteristic peaks: δ 9.29 (d, J=8.5 Hz, 1 H), 8.10 (s, 1 H), 7.04- 7.10 (m, 1 H), 7.00 (s, 1 H), 6.75 (s, 1 H), 5.05-5.30 (m, 3H), 4.00-4.07 (m, 1 H), 1 .42 (s, 3H), 1 .41 (s, 3H).

Step 4: Preparation of C104-Bis Na salt. A suspension of C104 (212 mg, 0.33 mmol) in water (10 mL) was cooled to 0 °C and treated with a solution of sodium bicarbonate (56.4 mg, 0.67 mmol) in water (2 mL), added dropwise. The reaction mixture was cooled to -70 °C (frozen) and lyophilized to afford C104-Bis Na salt as a white solid. Yield: 210 mg, 0.31 mmol, 93%. LCMS m/z 632.5 (M-1 ). 1H NMR (400 MHz, D20) δ 7.87 (s, 1 H), 6.94 (s, 1 H), 6.92 (s, 1 H), 5.35 (d, J=5 Hz, 1 H), 5.16 (s, 2H), 4.46-4.52 (m, 1 H), 3.71 (dd, half of ABX pattern, J=14.5, 6 Hz, 1 H), 3.55 (dd, half of ABX pattern, J=14.5, 6 Hz, 1 H), 1.43 (s, 3H), 1 .42 (s, 3H).

////////////Pfizer,  monobactam,  PF-?, 1380110-34-8, pfizer, pf, 1380110-45-1, WO 2012073138, Matthew Frank BrownSeungil HanManjinder LallMark. J. Mitton-FryMark Stephen PlummerHud Lawrence RisleyVeerabahu ShanmugasundaramJeremy T. Starr, preclinical

Abaloparatide, абалопаратид , أبالوباراتيد , 巴罗旁肽 ,

$
0
0

Chemical structure for Abaloparatide

Abaloparatide

BA058
BIM-44058
UNII-AVK0I6HY2U

BA058; BIM-44058; CAS  247062-33-5

MW 3960.5896, MF C174 H300 N56 O49

абалопаратид [Russian] [INN]
أبالوباراتيد [Arabic] [INN]
巴罗旁肽 [Chinese] [INN]
str1

NAME………C2.29-methyl(22-L-glutamic acid(F>E),23-L-leucine(F>L),25-L-glutamic acid(H>E),26-L-lysine(H>K),28-L-leucine(I>L),30-L-lysine(E>K),31-L-leucine(I>L))human parathyroid hormone-related protein-(1-34)-proteinamide
L-Alaninamide, L-alanyl-L-valyl-L-seryl-L-alpha-glutamyl-L-histidyl-L-glutaminyl-L-leucyl-L-leucyl-L-histidyl-L-alpha-aspartyl-L-lysylglycyl-L-lysyl-L-seryl-L-isoleucyl-L-glutaminyl-L-alpha-aspartyl-L-leucyl-L-arginyl-L-arginyl-L-arginyl-L-alpha-glutamyl-L-leucyl-L-leucyl-L-alpha-glutamyl-L-lysyl-L-leucyl-L-leucyl-2-methylalanyl-L-lysyl-L-leucyl-L-histidyl-L-threonyl-

L-Alaninamide, L-alanyl-L-valyl-L-seryl-L-α-glutamyl-L-histidyl-L-glutaminyl-L-leucyl-L-leucyl-L-histidyl-L-α-aspartyl-L-lysylglycyl-L-lysyl-L-seryl-L-isoleucyl-L-glutaminyl-L-α-aspartyl-L-leucyl-L-arginyl-L-arginyl-L-arginyl-L-α-glutamyl-L-leucyl-L-leucyl-L-α-glutamyl-L-lysyl-L-leucyl-L-leucyl-2-methylalanyl-L-lysyl-L-leucyl-L-histidyl-L-threonyl-

  1. C2.29-methyl(22-L-glutamic acid(F>E),23-L-leucine(F>L),25-L-glutamic acid(H>E),26-L-lysine(H>K),28-L-leucine(I>L),30-L-lysine(E>K),31-L-leucine(I>L))human parathyroid hormone-related protein-(1-34)-proteinamide

Biologic Depiction

Abaloparatide biologic depiction
IUPAC Condensed

H-Ala-Val-Ser-Glu-His-Gln-Leu-Leu-His-Asp-Lys-Gly-Lys-Ser-Ile-Gln-Asp-Leu-Arg-Arg-Arg-Glu-Leu-Leu-Glu-Lys-Leu-Leu-Aib-Lys-Leu-His-Thr-Ala-NH2

Sequence

AVSEHQLLHDKGKSIQDLRRRELLEKLLXKLHTA

HELM

PEPTIDE1{A.V.S.E.H.Q.L.L.H.D.K.G.K.S.I.Q.D.L.R.R.R.E.L.L.E.K.L.L.[Aib].K.L.H.T.A.[am]}$$$$

IUPAC

(N-(L-alanyl-L-valyl-L-seryl-L-alpha-glutamyl-L-histidyl-L-glutaminyl-L-leucyl-L-leucyl-L-histidyl-L-alpha-aspartyl-L-lysyl-glycyl-L-lysyl-L-seryl-L-isoleucyl-L-glutaminyl-L-alpha-aspartyl-L-leucyl-L-arginyl-L-arginyl-L-arginyl-L-alpha-glutamyl-L-leucyl-L-leucyl-L-alpha-glutamyl-L-lysyl-L-leucyl-L-leucyl)-2-aminoisobutyryl)-L-lysyl-L-leucyl-L-histidyl-L-threonyl-L-alaninamide

Tymlos

FDA 4/28/2017

To treat osteoporosis in postmenopausal women at high risk of fracture or those who have failed other therapies
Drug Trials Snapshot

2D chemical structure of 247062-33-5

Image result for AbaloparatideImage result for Abaloparatide

CLINICAL……….https://clinicaltrials.gov/search/intervention=Abaloparatide%20OR%20BA058%20OR%20BIM-44058

BIM-44058 is a 34 amino acid analog of native human PTHrP currently in phase III clinical trials at Radius Health for the treatment of postmenopausal osteoporosis. Radius is also developing a microneedle transdermal patch using a 3M drug delivery system in phase II clinical trials. The drug candidate was originally developed at Biomeasure (a subsidiary of Ipsen), and was subsequently licensed to Radius and Teijin Pharma.

Abaloparatide (brand name Tymlos; formerly BA058) is a parathyroid hormone-related protein (PTHrP) analog drug used to treat osteoporosis. Like the related drug teriparatide, and unlike bisphosphonates, it is an anabolic (i.e., bone growing) agent.[1] A subcutaneous injection formulation of the drug has completed a Phase III trial for osteoporosis.[2] This single study found a decrease in fractures.[3] In 28 April 2017, it was approved by Food and drug administration (FDA) to treat postmenopausal osteoporosis.

Image result for Abaloparatide

Therapeutics

Medical use

Abaloparatide is indicated to treat postmenopausal women with osteoporosis who are more susceptible to bone fractures.[2]

Dosage

The dose recommended is 80mcg subcutaneous injection once a day, administered in the periumbilical area using a prefilled pen device containing 30 doses.[4]

Warnings and Precautions

Preclinical studies revealed that abaloparatide systemic daily administration leads to a dose- and time-dependent increase in the incidence of osteosarcoma in rodents.[5] However, whether abaloparatide-SC will cause osteosarcoma in humans is unknown. Thus, the use of abaloparatide is not recommended for individuals at increased risk of osteosarcoma. Additionally, its use is not advised for more than 2 years during a patient’s lifetime.[4][6]

Image result for Abaloparatide

Side Effects

The most common side effects reported by more than 2% of clinical trials subjects are hypercalciuria, dizziness, nausea, headache, palpitations, fatigue, upper abdominal pain and vertigo.[4]

Pharmacology

Abaloparatide is 34 amino acid synthetic analog of PTHrP. It has 41% homology to parathyroid hormone (PTH) (1-34) and 76% homology to parathyroid hormone-related protein (PTHrP) (1-34).[7] It works as an anabolic agent for the bone, through selective activation of the parathyroid hormone 1 receptor (PTH1R), a G protein-coupled receptor (GPCR) expressed in the osteoblasts and osteocytes. Abaloparatide preferentially binds the RG conformational state of the PTH1R, which in turn elicits a transient downstream cyclic AMP signaling response towards to a more anabolic signaling pathway.[8][9]

History

Preclinical studies

Abaloropatide was previously known as BA058 and BIM-44058 while under development. The anabolic effects of abaloparatide on bone were demonstrated in two preclinical studies conducted in ovarectomized rats. Both studies showed increased cortical and trabecular bone volume and density, and trabecular microarchitecture improvement in vertebral and nonvertebral bones after short-term[10] and long-term[11] daily subcutaneous injection of abaloparatide compared to controls. Recent studies indicated a dose-dependent increased in bone mass and strength in long-term abalorapatide treatment.[12] However, it was also indicated that prolonged abalorapatide-SC treatment leads to increased incidence of osteosarcoma.[5] To date, there is no yet evidence for increased risk of bone tumors due to prolonged abalorapatide systemic administration in humans. Based on this preclinical data, the FDA does not advised the use of abaloparatide-SC for more than 2 years, or in patients with history of Paget disease and/or other conditions that exacerbates the risk of developing osteosarcoma.[4]

Clinical Trials

Phase II trials were initiated in 2008. A 24-week randomized trial was conducted in postmenopausal women with osteoporosis (n=222) assessing bone mass density (BMD) changes as the primary endpoint.[13] Significant BMD increase at doses of 40 and 80 mcg were found in the lumbar spine, femur and hips of abaloparatide-treated participants compared to placebo. Additionally, abaloparatide showed superior anabolic effects on the hips compared to teriparatide.[14]

In the phase III (2011-2014) Abaloparatide Comparator Trial in Vertebral Endpoints (ACTIVE) trial, a 18-months randomized, multicenter, double-blinded, placebo-controlled study evaluated the long-term efficacy of abaloparatide compared to placebo and teriparatide in 2,463 postmenopausal women (± 69 years old).[2] Women who received daily injections of abaloparatide experienced substantial reduction in the incidence of fractures compared to placebo. Additionally, greater BMD increase at 6, 12 and 18 months in spinal, hips and femoral bones was observed in abaloparatide compared to placebo and teriparatide-treated subjects.[3]

Participants who completed 18 months of abaloparatide or placebo in the ACTIVE study were invited to participate in an extended open-labeled study – ACTIVExtend study (2012-2016).[15] Subjects (n=1139) received additional 2 years of 70 mg of alendronate, Vitamin D (400 to 800 IU), and calcium (500–1000 mg) supplementation daily. Combined abaloparatide and alendronate therapy reduced significantly the incidence of vertebral and nonvertebral fractures.[16]

A clinical trial assessing the effectiveness of abaloparatide in altering spinal bone mineral density (BMD) in male subjects is expected to start in the first quarter of 2018. If successful, Radius Health aims to submit a sNDA to expand the use of abaloparatide-SC to treat men with osteoporosis.[17]

In addition to the injectable form of abaloparatide, a transdermal patch is also in development.[1]

Commercialization

As previously noted, abaloparatide-SC is manufactured by Radius Health, Inc. (Nasdaq: RDUS), a biomedical company based in Waltham, Massachusetts. This company is focused on the development of new therapeutics for osteoporosis, cancer and endocrine diseases. Abaloparatide is the only drug currently marketed by Radius Health. RDUS reported that sales for abaloparatide were $3.5million for the third quarter of 2017.[17] The company announced a net loss of $57.8 million, or $1.31 per share for the third quarter of 2017, compared to $19.2 million for the same quarter of 2016.[18] The net loss most likely reflects the substantial expenses associated with the preparation and launching of abaloparatide into the US market in May 2017.

In July 2017, Radius Health licensed rights to Teijin Limited for abaloparatide-SC manufacture and commercialization in Japan. Teijin is developing abaloparatide-SC under agreement with Ipsen Pharma S.A.S., and is conducting a phase III clinical trial in Japanese patients with osteoporosis.[19]

Regulatory Information

Radius Health filed a Marketing Authorization Application (MAA) in November 2015,[20] which was validated in December, 2015, and still under regulatory assessment by the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA). As in July 2017, the CHMP issued a second Day-180 List of Outstanding Issues, which Radius is addressing with the CHMP.[17]

In February 2016 a NDA was filed to the FDA, Radius NDA for abaloparatide-SC was accepted in May, 2016.[21] A Prescription Drug User Fee Act (PDUFA) date was initially granted in March 30, 2016, but then extended to June 30, 2017.[22] As previously stated, abaloparatide injection was approved for use in postmenopausal osteoporosis on April 28, 2017.[6]

Intellectual Property

Radius Health currently holds three patents on abaloparatide-SC, with expiration dates from 2027-2028.[23] The patents relate to the drug composition (US 8148333), and the drug delivery methods (US 7803770 B2 and US 8748382-B2).

As previously mentioned, Teijin Limited was granted use of Radius Health intellectual property in July 2017, for the development, manufacture and commercialization of abaloparatide-sc in Japan.

PATENT

http://www.google.com/patents/EP2206725A1?cl=en

  1. A peptide of the formula:

    [Glu22, 25, Leu23, 2831, Lys26, Aib29, Nle30]hPTHrP(1-34)NH2;
    [Glu22, 25, Leu23, 28, 3031, Lys26, Aib29]hPTHrP(1-34)NH2; [Glu22, 25,29, Leu23, 28, 30, 31, Lys26]hpTHrP(1-34)NH2; [Glu22, 25, 29, Leu23, 28, 31, Lys26, Nle30]hPTHrP(1-34)NH2; [Ser1, Ile5, Met8, Asn10, Leu11, 23, 28, 31, His14, Cha15, Glu22, 25, Lys26, 30, Aib29]hPTHrP (1-34)NH2; [Cha22, Leu23, 28, 31, Glu25, 29, Lys26, Nle30]hPTHrP(1-34)NH2; [Cha7, 1115]hPTHrP(1-34)NH2; [Cha7, 8, 15]hPTHrP(1-34)NH2; [Glu22, Leu23, 28, Aib25, 29, Lys26]hpTHrP(1-34)NH2; [Aib29]hPTHrP(1-34)NH2; [Glu22, 25, Leu23, 28, 31, Lys26, Aib29, 30]hPTHrP(1-34)NH2; [Glu22, 25, Leu23, 28, 31, Lys26, Aib29]hPTHrP(1-34)NH2; [Glu22, 25, Leu23, 28, 31, Aib26, 29, Lys30] hPTHrP(1-34)NH2; or [Leu27, Aib29]hPTH(1-34)NH2; or a pharmaceutically acceptable salt thereof.

PATENT

SEE……http://www.google.com.ar/patents/US8148333?cl=en

PATENT

SEE…………http://www.google.im/patents/US20090227498?cl=pt

EP5026436A Title not available
US3773919 Oct 8, 1970 Nov 20, 1973 Du Pont Polylactide-drug mixtures
US4767628 Jun 29, 1987 Aug 30, 1988 Imperial Chemical Industries Plc Polylactone and acid stable polypeptide
WO1994001460A1* Jul 13, 1993 Jan 20, 1994 Syntex Inc Analogs of pth and pthrp, their synthesis and use for the treatment of osteoporosis
WO1994015587A2 Jan 5, 1994 Jul 21, 1994 Steven A Jackson Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
WO1997002834A1* Jul 3, 1996 Jan 30, 1997 Biomeasure Inc Analogs of parathyroid hormone
WO1997002834A1* 3 Jul 1996 30 Jan 1997 Biomeasure Inc Analogs of parathyroid hormone
WO2008063279A2* 3 Oct 2007 29 May 2008 Radius Health Inc A stable composition comprising a bone anabolic protein, namely a pthrp analogue, and uses thereof
US5695955 * 23 May 1995 9 Dec 1997 Syntex (U.S.A.) Inc. Gene expressing a nucleotide sequence encoding a polypeptide for treating bone disorder
US20030166836 * 6 Nov 2002 4 Sep 2003 Societe De Conseils De Recherches Et D’application Scientefiques, S.A.S., A France Corporation Analogs of parathyroid hormone
US20050282749 * 14 Jan 2005 22 Dec 2005 Henriksen Dennis B Glucagon-like peptide-1 (GLP-1); immunotherapy; for treatment of obesity
Tymlos abaloparatide 4/28/2017 To treat osteoporosis in postmenopausal women at high risk of fracture or those who have failed other therapies
Drug Trials Snapshot
Abaloparatide
Clinical data
Trade names Tymlos
Synonyms BA058, BIM-44058
Routes of
administration
Subcutaneous injection
ATC code
  • none
Legal status
Legal status
  • Investigational
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
Chemical and physical data
Formula C174H299N56O49
Molar mass 3,959.65 g·mol−1
3D model (JSmol)

/////////FDA 2017, Abaloparatide, TYMLOS, RADIUS HEALTH, PEPTIDE, BA058, BIM 44058; 247062-33-5, абалопаратид أبالوباراتيد 巴罗旁肽 

CCC(C)C(C(=O)NC(CCC(=O)N)C(=O)NC(CC(=O)O)C(=O)NC(CC(C)C)C(=O)NC(CCCNC(=N)N)C(=O)NC(CCCNC(=N)N)C(=O)NC(CCCNC(=N)N)C(=O)NC(CCC(=O)O)C(=O)NC(CC(C)C)C(=O)NC(CC(C)C)C(=O)NC(CCC(=O)O)C(=O)NC(CCCCN)C(=O)NC(CC(C)C)C(=O)NC(CC(C)C)C(=O)NC(C)(C)C(=O)NC(CCCCN)C(=O)NC(CC(C)C)C(=O)NC(CC1=CN=CN1)C(=O)NC(C(C)O)C(=O)NC(C)C(=O)N)NC(=O)C(CO)NC(=O)C(CCCCN)NC(=O)CNC(=O)C(CCCCN)NC(=O)C(CC(=O)O)NC(=O)C(CC2=CN=CN2)NC(=O)C(CC(C)C)NC(=O)C(CC(C)C)NC(=O)C(CCC(=O)N)NC(=O)C(CC3=CN=CN3)NC(=O)C(CCC(=O)O)NC(=O)C(CO)NC(=O)C(C(C)C)NC(=O)C(C)N

Viewing all 1676 articles
Browse latest View live